1
|
August I, Gagneux P, Semendeferi K, Marchetto MC. Evolution of Human Susceptibility to Alzheimer's Disease: A Review of Hypotheses and Comparative Evidence. Evol Anthropol 2025; 34:e22054. [PMID: 39806778 DOI: 10.1002/evan.22054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025]
Abstract
Primates rely on memory to navigate both physical and social environments and in humans, loss of memory function leads to devastating consequences. Alzheimer's disease (AD) is a neurodegenerative disease which begins by impacting memory functioning and is ultimately fatal. AD is common across human populations and its prevalence is predicted to rise with increases in the aging population. Despite this, the full AD phenotype has not been observed in any other nonhuman primate species. While a significant amount of research has been devoted to understanding the immediate mechanisms involved in AD pathogenesis in humans, less research has focused on why humans are particularly vulnerable to neurodegenerative diseases like AD. Here we explore hypotheses on the evolution of distinct human susceptibility to AD and place these in the context of findings from comparative neuroanatomical and molecular studies and discuss recent evidence for evolutionary changes protective against AD in the primate lineage.
Collapse
Affiliation(s)
- Isabel August
- Department of Anthropology, University of California San Diego, La Jolla, California, USA
| | - Pascal Gagneux
- Department of Anthropology, University of California San Diego, La Jolla, California, USA
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, California, USA
| | - Katerina Semendeferi
- Department of Anthropology, University of California San Diego, La Jolla, California, USA
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, California, USA
| | - Maria Carolina Marchetto
- Department of Anthropology, University of California San Diego, La Jolla, California, USA
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, California, USA
| |
Collapse
|
2
|
Zhu X, Sun S, Lin L, Wu Y, Ma X. Transformer-based approaches for neuroimaging: an in-depth review of their role in classification and regression tasks. Rev Neurosci 2025; 36:209-228. [PMID: 39333087 DOI: 10.1515/revneuro-2024-0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/13/2024] [Indexed: 09/29/2024]
Abstract
In the ever-evolving landscape of deep learning (DL), the transformer model emerges as a formidable neural network architecture, gaining significant traction in neuroimaging-based classification and regression tasks. This paper presents an extensive examination of transformer's application in neuroimaging, surveying recent literature to elucidate its current status and research advancement. Commencing with an exposition on the fundamental principles and structures of the transformer model and its variants, this review navigates through the methodologies and experimental findings pertaining to their utilization in neuroimage classification and regression tasks. We highlight the transformer model's prowess in neuroimaging, showcasing its exceptional performance in classification endeavors while also showcasing its burgeoning potential in regression tasks. Concluding with an assessment of prevailing challenges and future trajectories, this paper proffers insights into prospective research directions. By elucidating the current landscape and envisaging future trends, this review enhances comprehension of transformer's role in neuroimaging tasks, furnishing valuable guidance for further inquiry.
Collapse
Affiliation(s)
- Xinyu Zhu
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| | - Shen Sun
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| | - Lan Lin
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| | - Yutong Wu
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| | - Xiangge Ma
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| |
Collapse
|
3
|
Kedia S, Simons M. Oligodendrocytes in Alzheimer's disease pathophysiology. Nat Neurosci 2025:10.1038/s41593-025-01873-x. [PMID: 39881195 DOI: 10.1038/s41593-025-01873-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
Our understanding of Alzheimer's disease (AD) has transformed from a purely neuronal perspective to one that acknowledges the involvement of glial cells. Despite remarkable progress in unraveling the biology of microglia, astrocytes and vascular elements, the exploration of oligodendrocytes in AD is still in its early stages. Contrary to the traditional notion of oligodendrocytes as passive bystanders in AD pathology, emerging evidence indicates their active participation in and reaction to amyloid and tau pathology. Oligodendrocytes undergo a functional transition to a disease-associated state, engaging in immune modulation, stress responses and cellular survival. Far from being inert players, they appear to serve a dual role in AD pathogenesis, potentially offering defense mechanisms against pathology while also contributing to disease progression. This Review explores recent advancements in understanding the roles of oligodendrocytes and their myelin sheaths in the context of AD, shedding light on their complex interactions within the disease pathology.
Collapse
Affiliation(s)
- Shreeya Kedia
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
4
|
Ma X, Wang XM, Tang GZ, Wang Y, Liu XC, Wang SD, Peng P, Qi XH, Qin XY, Wang YJ, Wang CW, Zhou JN. Alterations of amino acids in older adults with Alzheimer's Disease and Vascular Dementia. Amino Acids 2025; 57:10. [PMID: 39825947 PMCID: PMC11742867 DOI: 10.1007/s00726-024-03442-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/31/2024] [Indexed: 01/20/2025]
Abstract
Metabolomics provide a promising tool for understanding dementia pathogenesis and identifying novel biomarkers. This study aimed to identify amino acid biomarkers for Alzheimer's Disease (AD) and Vascular Dementia (VD). By amino acid metabolomics, the concentrations of amino acids were determined in the serum of AD and VD patients as well as age-matched healthy controls. Several differences in the concentration of amino acids were observed in AD patients compared to both healthy controls and VD patients. However, no significant distinction was found between healthy controls and VD patients. Considering comorbidities, cystine levels were higher in AD than in VD among non-diabetic patients, but not in those with diabetes. Notably, creatine, spermidine, cystine, and tyrosine demonstrated favorable results in decision curve analyses and good discriminative performances, suggesting their potential for clinical application. These fundings give novel perspectives of serum amino acids for predicting metabolic pathways in AD and VD pathogenesis.
Collapse
Affiliation(s)
- Xin Ma
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
- Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Xin-Meng Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
| | - Guo-Zhang Tang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
- Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Yi Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
- First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Xue-Chun Liu
- Department of Neurology, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, P. R. China
| | - Shuai-Deng Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Peng Peng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
- First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Xiu-Hong Qi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, 230026, P. R. China
| | - Xin-Ya Qin
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, 230026, P. R. China
- Institute of Brain Science, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
| | - Yue-Ju Wang
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, P. R. China.
| | - Chen-Wei Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, P. R. China.
| | - Jiang-Ning Zhou
- Institute of Brain Science, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
| |
Collapse
|
5
|
Li H, Zhao H, Chen L, Zhang W, Zhang W, Cheng X. Rapid screening of acetylcholinesterase inhibitors in Qi-Fu-Yin using magnetic metal-organic frameworks immobilized with acetylcholinesterase. Bioorg Chem 2025; 156:108183. [PMID: 39827651 DOI: 10.1016/j.bioorg.2025.108183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Current immobilization approaches for ligand fishing often experience challenges such as limited protein loading capacity and difficulties in the recycling process. To overcome these challenges, we synthesized a magnetic metal-organic frameworks (MMOFs) composite, which can be rapidly separated and has a large specific surface area, and employed it to immobilize acetylcholinesterase (AChE). The synthesized MMOFs@AChE composite exhibited a high immobilization yield (129.7 mg/g) and excellent relative activity recovery (88.1 %). Furthermore, immobilized AChE can improve its resistance to alkaline environments and high temperatures. After being stored at 4 °C for a month, the immobilized enzyme maintained 91.4 % of its original activity, significantly higher than the free enzyme (77.6 %). Furthermore, it preserved more than 80 % of its initial activity after five cycles and 68.7 % after eight cycles. The composite MMOFs@AChE was then incubated with Qi-Fu-Yin extract to fish for ligands binding to AChE. Notably, Qi-Fu-Yin can alleviate Alzheimer's disease (AD) symptoms by modulating the AChE pathway, while active compounds remain unclear. Sixteen potential AChE inhibitors were identified through UHPLC-Q-Exactive-Orbitrap-MS/MS. The results of ligand fishing were validated through molecular docking studies, molecular dynamics simulation, surface plasmon resonance and AChE inhibitory activity assays. The screened compounds may exert inhibitory effects on AChE by altering the spatial configuration of the catalytic site or by influencing the binding of the substrate to the catalytic site, catalytic anionic site and peripheral anionic site regions. The MMOFs@AChE-based ligand fishing platform offers an efficient, effective, and convenient approach for enzymatic inhibitors discovery from natural products.
Collapse
Affiliation(s)
- Hengyu Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078 China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China.
| | - Hongwei Zhao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China
| | - Lingxiao Chen
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China
| | - Wenting Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China
| | - Wei Zhang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078 China.
| | - Xiaorui Cheng
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078 China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355 China.
| |
Collapse
|
6
|
Markovich SW, Frey BL, Scalf M, Shortreed MR, Smith LM. Dehydroamino acids and their crosslinks in Alzheimer's disease aggregates. Brain Commun 2025; 7:fcaf019. [PMID: 39882022 PMCID: PMC11775630 DOI: 10.1093/braincomms/fcaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/16/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of protein aggregates, which are thought to be influenced by posttranslational modifications (PTMs). Dehydroamino acids (DHAAs) are rarely observed PTMs that contain an electrophilic alkene capable of forming protein-protein crosslinks, which may lead to protein aggregation. We report here the discovery of DHAAs in the protein aggregates from AD, constituting an unknown and previously unsuspected source of extensive proteomic complexity. We used mass spectrometry-based proteomics to discover 404 sites of DHAA formation in 171 proteins from protein aggregate-enriched human brain samples, 6-fold more sites than observed in the soluble protein fractions. The DHAA modifications are observed both directly and in the form of conjugates after reacting with abundant cellular nucleophiles or crosslinking to nucleophilic amino acid residues. We report 11 such crosslinks, including three in the Tau protein, which are 10-fold more abundant in AD samples compared with age-matched controls. Many of the proteins found to contain DHAAs and their conjugates are involved in protein aggregation or pathways dysregulated in AD. DHAAs are prevalent modifications in the AD brain proteome and give rise to protein crosslinks that may contribute to protein aggregation.
Collapse
Affiliation(s)
- Samuel W Markovich
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| | - Brian L Frey
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| | - Mark Scalf
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| | - Michael R Shortreed
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| | - Lloyd M Smith
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| |
Collapse
|
7
|
Kuziak A, Heczko P, Pietrzyk A, Strus M. Iron Homeostasis Dysregulation, Oro-Gastrointestinal Microbial Inflammatory Factors, and Alzheimer's Disease: A Narrative Review. Microorganisms 2025; 13:122. [PMID: 39858890 PMCID: PMC11767265 DOI: 10.3390/microorganisms13010122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/23/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, is a progressive neurodegenerative disorder that profoundly impacts cognitive function and the nervous system. Emerging evidence highlights the pivotal roles of iron homeostasis dysregulation and microbial inflammatory factors in the oral and gut microbiome as potential contributors to the pathogenesis of AD. Iron homeostasis disruption can result in excessive intracellular iron accumulation, promoting the generation of reactive oxygen species (ROS) and oxidative damage. Additionally, inflammatory agents produced by pathogenic bacteria may enter the body via two primary pathways: directly through the gut or indirectly via the oral cavity, entering the bloodstream and reaching the brain. This infiltration disrupts cellular homeostasis, induces neuroinflammation, and exacerbates AD-related pathology. Addressing these mechanisms through personalized treatment strategies that target the underlying causes of AD could play a critical role in preventing its onset and progression.
Collapse
Affiliation(s)
- Agata Kuziak
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, św. Łazarza 16 Street, 31-008 Cracow, Poland;
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| | - Piotr Heczko
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| | - Agata Pietrzyk
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| | - Magdalena Strus
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| |
Collapse
|
8
|
Dickerson BC, Atri A, Clevenger C, Karlawish J, Knopman D, Lin P, Norman M, Onyike C, Sano M, Scanland S, Carrillo M. The Alzheimer's Association clinical practice guideline for the Diagnostic Evaluation, Testing, Counseling, and Disclosure of Suspected Alzheimer's Disease and Related Disorders (DETeCD-ADRD): Executive summary of recommendations for specialty care. Alzheimers Dement 2025; 21:e14337. [PMID: 39713957 PMCID: PMC11772716 DOI: 10.1002/alz.14337] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 12/24/2024]
Abstract
US clinical practice guidelines for the diagnostic evaluation of cognitive impairment due to Alzheimer's disease (AD) or a related dementia (ADRD) are two decades old. This evidence-based guideline was developed to empower all clinicians to implement a structured approach for evaluating a patient with symptoms that may represent clinical AD/ADRD. An expert workgroup conducted a review of 7374 publications (133 met inclusion criteria) and developed recommendations as steps in an evaluation process. This summary briefly reviews core recommendations and details specialist recommendations of a high-quality, evidence-supported evaluation process aimed at characterizing, diagnosing, and disclosing the patient's cognitive functional status, cognitive-behavioral syndrome, and likely underlying brain disease so that optimal care plans to maximize patient/care partner dyad quality of life can be developed; a companion article summarizes primary care recommendations. If clinicians use the recommendations in this guideline and health-care systems provide adequate resources, outcomes should improve in most patients in most practice settings. HIGHLIGHTS: US clinical practice guidelines for the diagnostic evaluation of cognitive impairment due to Alzheimer's disease (AD) or related dementias (ADRD) are decades old and aimed at specialists. This evidence-based guideline was developed to empower all-including primary care-clinicians to implement a structured approach for evaluating a patient with symptoms that may represent clinical AD/ADRD. This summary focuses on recommendations appropriate for specialty practice settings, forming key elements of a high-quality, evidence-supported evaluation process aimed at characterizing, diagnosing, and disclosing the patient's cognitive functional status, cognitive-behavioral syndrome, and likely underlying brain disease so that optimal care plans to maximize patient/care partner dyad quality of life can be developed; a companion article summarizes primary care recommendations. If clinicians use this guideline and health-care systems provide adequate resources, outcomes should improve in most patients in most practice settings.
Collapse
Affiliation(s)
- Bradford C. Dickerson
- Frontotemporal Disorders Unit, Department of NeurologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Alireza Atri
- Banner Sun Health Research Institute and Banner Alzheimer's InstituteSun CityArizonaUSA
- Department of NeurologyCenter for Brain/Mind MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Carolyn Clevenger
- Department of Neurology, Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Jason Karlawish
- Departments of Medicine, Medical Ethics and Health Policy, and Neurology, Perelman School of Medicine, Penn Memory CenterUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - David Knopman
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Pei‐Jung Lin
- Center for the Evaluation of Value and Risk in HealthInstitute for Clinical Research and Health Policy Studies, Tufts Medical CenterBostonMassachusettsUSA
| | - Mary Norman
- Cedars‐Sinai Medical CenterCulver CityCaliforniaUSA
| | - Chiadi Onyike
- Division of Geriatric Psychiatry and NeuropsychiatryThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Mary Sano
- James J. Peters VAMCBronxNew YorkUSA
- Department of PsychiatryAlzheimer's Disease Research CenterIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Maria Carrillo
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| |
Collapse
|
9
|
Pini L, Lista S, Griffa A, Allali G, Imbimbo BP. Can brain network connectivity facilitate the clinical development of disease-modifying anti-Alzheimer drugs? Brain Commun 2024; 7:fcae460. [PMID: 39741782 PMCID: PMC11686405 DOI: 10.1093/braincomms/fcae460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025] Open
Abstract
The preclinical phase of Alzheimer's disease represents a crucial time window for therapeutic intervention but requires the identification of clinically relevant biomarkers that are sensitive to the effects of disease-modifying drugs. Amyloid peptide and tau proteins, the main histological hallmarks of Alzheimer's disease, have been widely used as biomarkers of anti-amyloid and anti-tau drugs. However, these biomarkers do not fully capture the multiple biological pathways of the brain. Indeed, robust amyloid-target engagement by anti-amyloid monoclonal antibodies has recently translated into modest cognitive and clinical benefits in Alzheimer's disease patients, albeit with potentially life-threatening side effects. Moreover, targeting the tau pathway has yet to result in any positive clinical outcomes. Findings from computational neuroscience have demonstrated that brain regions do not work in isolation but are interconnected within complex network structures. Brain connectivity studies suggest that misfolded proteins can spread through these connections, leading to the hypothesis that Alzheimer's disease is a pathology of network disconnectivity. Based on these assumptions, here we discuss how incorporating brain connectivity outcomes could better capture global brain functionality and, in conjunction with traditional Alzheimer's disease biomarkers, could facilitate the clinical development of new disease-modifying anti-Alzheimer's disease drugs.
Collapse
Affiliation(s)
- Lorenzo Pini
- Department of Neuroscience, Università degli Studi di Padova, 35121 Padova, Italy
- Padova Neuroscience Center, Università degli Studi di Padova, 35121 Padova, Italy
| | - Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain
| | - Alessandra Griffa
- Department of Clinical Neurosciences, Leenaards Memory Center, Lausanne University Hospital and University of Lausanne, Montpaisible 16, 1011 Lausanne, Switzerland
- Medical Image Processing Laboratory, Neuro-X Institute, École Polytechnique Fédérale De Lausanne (EPFL), Campus Biotech Chemin des Mines 9, 1202 Geneva, Switzerland
| | - Gilles Allali
- Department of Clinical Neurosciences, Leenaards Memory Center, Lausanne University Hospital and University of Lausanne, Montpaisible 16, 1011 Lausanne, Switzerland
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, 43122 Parma, Italy
| |
Collapse
|
10
|
Engelhardt E, Resende EDPF, Gomes KB. Physiopathological mechanisms underlying Alzheimer's disease: a narrative review. Dement Neuropsychol 2024; 18:e2024VR01. [PMID: 39697643 PMCID: PMC11654088 DOI: 10.1590/1980-5764-dn-2024-vr01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 12/20/2024] Open
Abstract
The neuropathological signature of Alzheimer's disease (AD) comprises mainly amyloid plaques, and neurofibrillary tangles, resulting in synaptic and neuronal loss. These pathological structures stem from amyloid dysfunctional metabolism according to the amyloid cascade hypothesis, leading to the formation of plaques, and apparently inducing the initiation of the abnormal tau pathway, with phosphorylation and aggregation of these proteins, ultimately causing the formation of tangles. In this narrative review, the existing hypothesis related to the pathophysiology of AD were compiled, and biological pathways were highlighted in order to identify the molecules that could represent biological markers of the disease, necessary to establish early diagnosis, as well as the selection of patients for therapeutical interventional strategies.
Collapse
Affiliation(s)
- Eliasz Engelhardt
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo Horizonte MG, Brazil
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte MG, Brazil
| | - Karina Braga Gomes
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Belo Horizonte MG, Brazil
| |
Collapse
|
11
|
Zeng X, Sheng Z, Zhang Y, Xiao J, Li Y, Zhang J, Xu G, Jia J, Wang M, Li L. The therapeutic potential of glycyrrhizic acid and its metabolites in neurodegenerative diseases: Evidence from animal models. Eur J Pharmacol 2024; 985:177098. [PMID: 39510337 DOI: 10.1016/j.ejphar.2024.177098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Neurodegenerative diseases, mostly occurring in the elderly population, are the significant cause of disability and death worldwide. The pathogenesis of neurodegenerative diseases is still largely unknown yet, although they have been continuously explored. Thus, there is still a lack of safe, effective, and low side effect drugs in clinical practice for the treatment of neurodegenerative diseases. Pieces of accumulating evidence have demonstrated that licorice played neuroprotective roles in various neurodegenerative diseases. In the past two decades, increasing studies have indicated that glycyrrhizic acid (GL), the main active ingredient from traditional Chinese medicine licorice (widely used in the food industry) and a triterpenoid saponin with multiple pharmacological effects (such as anti-oxidant, anti-inflammatory, and immune regulation), and its metabolites (glycyrrhetinic acid and carbenoxolone) play a neuroprotective role in a range of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease and epilepsy. This review will elaborate on the multiple neuroprotective mechanisms of GL and its metabolites in this series of diseases, aiming to provide a basis for further research on these protective drugs for neurodegenerative diseases and their clinical application. In summary, GL may be a promising candidate drug for the therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiansi Zeng
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, 314001, China; Department of Biochemistry and Molecular Biology, Jiaxing University Medical College, Jiaxing, 314001, China; Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, China
| | - Zixuan Sheng
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, 314001, China
| | - Yuqian Zhang
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, 314001, China
| | - Jing Xiao
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, 314001, China
| | - Yang Li
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, 314001, China
| | - Jiaping Zhang
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, 314001, China
| | - Guangtao Xu
- Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, China
| | - Jinjing Jia
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, 314001, China; Department of Physiology, Jiaxing University Medical College, Jiaxing, 314001, China.
| | - Min Wang
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, 314001, China; Department of Physiology, Jiaxing University Medical College, Jiaxing, 314001, China.
| | - Li Li
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, 314001, China; Department of Physiology, Jiaxing University Medical College, Jiaxing, 314001, China.
| |
Collapse
|
12
|
Elbatrawy AA, Ademoye TA, Alnakhala H, Tripathi A, Plascencia-Villa G, Zhu X, Perry G, Dettmer U, Fortin JS. Exploring the rhodanine universe: Design and synthesis of fluorescent rhodanine-based derivatives as anti-fibrillar and anti-oligomer agents against α-synuclein and 2N4R tau. Bioorg Med Chem 2024; 116:117990. [PMID: 39550891 PMCID: PMC11645236 DOI: 10.1016/j.bmc.2024.117990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
Tau and α-synuclein (α-syn) are prone-to-aggregate proteins that can be responsible for pathological lesions found in the brains of Alzheimer's disease (AD), Lewy body dementia (LBD), and Parkinson's disease (PD) patients. The early-stage oligomers and protofibrils of tau are believed to be strongly linked to human cognitive impairment while the toxic α-syn oligomers are associated with behavioral motor deficits. Therefore, concurrent targeting of both proteinaceous aggregates and oligomers are very challenging. Herein, rhodanine-based compounds were designed and synthesized to target the fibrils and oligomers of tau and α-syn proteins. In particular, the indole-containing rhodanines 5l and 5r displayed significantly high anti-aggregation activity towards α-syn fibrils by reducing of the thioflavin-T (ThT) fluorescence to less than 5 %. Moreover, 5r showed a remarkable decrease in the fluorescence of thioflavin-S (ThS) when incubated with the non-phosphorylated tau 0N4R and 2N4R, as well as the hyperphosphorylated tau isoform 1N4R. Transmission electron microscopy (TEM) analyses validated the powerful anti-fibrillar activity of 5l and 5r towards both protein aggregates. In addition, both 5l and 5r highly suppressed 0N4R tau and α-syn oligomer formation using the photo-induced cross-linking of unmodified protein (PICUP) assay. The fluorescence emission intensity of 5l was quenched to almost half in the presence of both protein fibrils at 510 nm. 5r showed a similar fluorescence response upon binding to 2N4R fibrils while no quenching effect was observed with α-syn aggregates. Ex vivo disaggregation assay using extracted human Aβ plaques was employed to confirm the ability of 5l and 5r to disaggregate the dense fibrils. Both inhibitors reduced the Aβ fibrils isolated from AD brains. 5l and 5r failed to show activity toward the cell-based α-syn inclusion formation. However, another indolyl derivative 5j prevented the α-syn inclusion at 5 µM. Collectively, the indolyl-rhodanine scaffold could act as a building block for further structural optimization to obtain dual targeting disease-modifying candidates for AD, LBD, and PD.
Collapse
Affiliation(s)
- Ahmed A Elbatrawy
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, United States
| | - Taiwo A Ademoye
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, United States
| | - Heba Alnakhala
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Arati Tripathi
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Germán Plascencia-Villa
- Deptartment of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, United States
| | - George Perry
- Deptartment of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Jessica S Fortin
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
13
|
Jiao JJ, Hu Y, Cui YJ, Tuo CM, Wang YX, Li XY, Zhang Y, Wu MN. Anisomycin alleviates cognitive impairments and pathological features in 3xTg-AD mice. Neuropharmacology 2024; 261:110159. [PMID: 39303856 DOI: 10.1016/j.neuropharm.2024.110159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Neuroinflammation plays a pivotal role in the pathogenesis of Alzheimer's disease (AD). Anisomycin is a pyrrolidine antibiotic isolated from Streptomyces griseolus, which is an efficient anti-inflammatory agent that functions both in vivo and in vitro. However, it is not clear whether anisomycin can exert neuroprotective effect in AD. In the present study, anisomycin was intragastrically administrated to female triple-transgenic AD (3xTg-AD) model mice, then Morris water maze test was used to observe the long-term spatial memory of mice, the in vivo hippocampal field potential recording was performed to evaluate the synaptic plasticity, the Western blot and immunofluorescence were employed to detect pathological changes, and the bioinformatics analysis was used to predict the potential target of anisomycin exerting effects in AD. The results showed that anisomycin ameliorated the long-term spatial memory deficits, improved LTP depression and increased the expression of PSD-95, reduced the Aβ and tau pathologies, and alleviated the activation of microglia and astrocytes in the brains of 3xTg-AD mice. In addition, the results from bioinformatics analysis showed that the potential target of anisomycin focused on inflammatory pathway. These results indicated that anisomycin exerts neuroprotective effects in 3xTg-AD mice by alleviating neuroinflammation, but the potential mechanism of anisomycin exerting neuroprotective effects needs to be further investigated.
Collapse
Affiliation(s)
- Juan-Juan Jiao
- Department of Physiology Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, China
| | - Yang Hu
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Yu-Jia Cui
- Department of Physiology Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, China
| | - Chun-Mei Tuo
- Department of Physiology Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, China
| | - Yi-Xuan Wang
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Xin-Yi Li
- Department of Neurology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China.
| | - Mei-Na Wu
- Department of Physiology Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology in Shanxi Province, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
14
|
Fu XX, Huang ZH, Wang SY, Qi JW, Luo ZJ, E Y, Zhang YD, Jiang T. Knockdown of TREML2 Alleviates Neuropathological Hallmarks and Cognitive Deficiency in a Model of Sporadic Alzheimer's Disease. J Inflamm Res 2024; 17:10471-10478. [PMID: 39654855 PMCID: PMC11627106 DOI: 10.2147/jir.s489474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024] Open
Abstract
Objective Recently, we revealed that triggering receptor expressed on myeloid cells-like 2 (TREML2) modulated inflammation by regulating microglial polarization and NLRP3 inflammasome activation. However, the role of TREML2 in Alzheimer's disease (AD) pathogenesis remains poorly understood. In this study, we tried to observe the impact of TREML2 on neuropathological hallmarks (including amyloid-β (Aβ) pathology, hyperphosphorylated tau and neuroinflammation) and cognitive deficiency in senescence-accelerated mouse prone substrain 8 (SAMP8) mice, an animal model of sporadic AD. Methods A lentiviral-based strategy was employed to manipulate TREML2 levels in the brain of SAMP8 mice. Enzyme-linked immunosorbent assay was used to detect the protein levels of inflammatory cytokines, Aβ42 and hyperphosphorylated tau. The mRNA levels of microglial polarization markers were assessed by qRT-PCR. Morris water maze test was performed to evaluate the spatial cognitive functions. Results TREML2 overexpression elevated inflammatory cytokines levels, induced microglial M1-type polarization, and exacerbated Aβ and tau pathology in SAMP8 mice. Contrastingly, knocking down TREML2 mitigated inflammatory cytokines release, promoted microglial M2-type polarization, ameliorated Aβ and tau pathology, and rescued cognitive deficiency in SAMP8 mice. Conclusion This study offers the first in vivo evidence that TREML2 contributes to the pathogenesis of AD. Furthermore, this study also proves that inhibition of TREML2 signaling may represent a potential treatment strategy for this disease.
Collapse
Affiliation(s)
- Xin-Xin Fu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Pharmacology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Zhi-Hang Huang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Shi-Yao Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Jing-Wen Qi
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Zi-Jian Luo
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yan E
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Ying-Dong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Teng Jiang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
15
|
Dubois B, Villain N, Schneider L, Fox N, Campbell N, Galasko D, Kivipelto M, Jessen F, Hanseeuw B, Boada M, Barkhof F, Nordberg A, Froelich L, Waldemar G, Frederiksen KS, Padovani A, Planche V, Rowe C, Bejanin A, Ibanez A, Cappa S, Caramelli P, Nitrini R, Allegri R, Slachevsky A, de Souza LC, Bozoki A, Widera E, Blennow K, Ritchie C, Agronin M, Lopera F, Delano-Wood L, Bombois S, Levy R, Thambisetty M, Georges J, Jones DT, Lavretsky H, Schott J, Gatchel J, Swantek S, Newhouse P, Feldman HH, Frisoni GB. Alzheimer Disease as a Clinical-Biological Construct-An International Working Group Recommendation. JAMA Neurol 2024; 81:1304-1311. [PMID: 39483064 DOI: 10.1001/jamaneurol.2024.3770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Importance Since 2018, a movement has emerged to define Alzheimer disease (AD) as a purely biological entity based on biomarker findings. The recent revision of the Alzheimer's Association (AA) criteria for AD furthers this direction. However, concerns about a purely biological definition of AD being applied clinically, the understanding of AD by society at large, and the translation of blood-based biomarkers into clinical practice prompt these International Working Group (IWG) updated recommendations. Objective To consider the revised AA criteria and to offer an alternative definitional view of AD as a clinical-biological construct for clinical use. The recommendations of the 2021 IWG diagnostic criteria are updated for further elaborating at-risk and presymptomatic states. Evidence Review PubMed was searched for articles published between July 1, 2020, and March 1, 2024, using the terms "biomarker" OR "amyloid" OR "tau" OR "neurodegeneration" OR "preclinical" OR "CSF" OR "PET" OR "plasma" AND "Alzheimer's disease." The references of relevant articles were also searched. Findings In the new AA diagnostic criteria, AD can be defined clinically as encompassing cognitively normal people having a core 1 AD biomarker. However, recent literature shows that the majority of biomarker-positive cognitively normal individuals will not become symptomatic along a proximate timeline. In the clinical setting, disclosing a diagnosis of AD to cognitively normal people with only core 1 AD biomarkers represents the most problematic implication of a purely biological definition of the disease. Conclusions and Relevance The ultimate aim of the field was to foster effective AD treatments, including preventing symptoms and dementia. The approach of diagnosing AD without a clinical and biological construct would be unwarranted and potentially concerning without a clear knowledge of when or whether symptoms will ever develop. It is recommended that those who are amyloid-positive only and, more generally, most biomarker-positive cognitively normal individuals, should not be labeled as having AD. Rather, they should be considered as being at risk for AD. The expansion of presymptomatic AD is viewed as a better diagnostic construct for those with a specific pattern of biomarkers, indicating that they are proximate to the expression of symptoms in the near future.
Collapse
Affiliation(s)
- Bruno Dubois
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Institute of Memory and Alzheimer's Disease, Paris, France
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau - ICM, FrontLab, Paris, France
| | - Nicolas Villain
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Institute of Memory and Alzheimer's Disease, Paris, France
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau - ICM, Maladie d'Alzheimer, Maladies à Prions, Paris, France
| | - Lon Schneider
- Keck School of Medicine of the University of Southern California, Los Angeles
| | - Nick Fox
- Department of Neurodegenerative Disease, Dementia Research Centre, and the United Kingdom Dementia Research Institute, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Noll Campbell
- Purdue University College of Pharmacy, West Lafayette, Indiana
- Purdue University Center for Aging and the Life Course, West Lafayette, Indiana
- Indiana University Center for Aging Research, Indianapolis
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla
| | - Miia Kivipelto
- Center for Alzheimer Research, Karolinska Institutet, Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Frank Jessen
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases, Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Bernard Hanseeuw
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Institute of Neurosciences, UC Louvain, Brussels, Belgium
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Frederik Barkhof
- Centre for Medical Image Computing, Department of Medical Physics and Bioengineering, University College London, London, United Kingdom
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, The Aging Brain, Karolinska University Hospital, Stockholm, Sweden
| | - Lutz Froelich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Steen Frederiksen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alessandro Padovani
- Neurology and Neurophysiology Section, Department Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Hospital Department of Continuità di Cura e Fragilità, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Vincent Planche
- Univ. Bordeaux, CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- Pôle de Neurosciences Cliniques, Centre Mémoire de Ressources et de Recherche, CHU de Bordeaux, Bordeaux, France
| | - Christopher Rowe
- Department of Molecular Imaging and Therapy, Austin Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Agustin Ibanez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Stefano Cappa
- University School for Advanced Studies, Pavia, Italy
- RCCS Mondino Foundation, Pavia, Italy
| | - Paulo Caramelli
- Behavioral and Cognitive Neurology Unit, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Nitrini
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ricardo Allegri
- Department of Cognitive Neurology, Fleni Neurological Institute, Buenos Aires, Argentina
- Department of Cognitive Neurosciences, Universidad de la Costa, Barranquilla, Colombia
| | - Andrea Slachevsky
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Memory and Neuropsychiatric Center Neurology Department, Hospital del Salvador and Neuropsychology and Clinical Neuroscience Laboratory, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Chile
- Neurology and Psychiatry Department, Clínica Alemana-Universidad Desarrollo, Santiago, Chile
| | - Leonardo Cruz de Souza
- Behavioral and Cognitive Neurology Unit, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andrea Bozoki
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill
| | - Eric Widera
- Division of Geriatrics, University of California, San Francisco
- Hospice & Palliative Care, San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Craig Ritchie
- Brain Health and Neurodegenerative Medicine, University of St Andrews, Scotland, United Kingdom
- Scottish Brain Sciences, Edinburgh, Scotland, United Kingdom
| | - Marc Agronin
- Medical Office for MIND Institute, Miami, Florida
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Lisa Delano-Wood
- Veterans Affairs San Diego Healthcare System, San Diego, California
- Department of Psychiatry, University of California, San Diego Health, La Jolla, California
- Center for Stress and Mental Health, VA San Diego Healthcare System, San Diego, California
| | - Stéphanie Bombois
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Institute of Memory and Alzheimer's Disease, Paris, France
| | - Richard Levy
- Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Institute of Memory and Alzheimer's Disease, Paris, France
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau - ICM, FrontLab, Paris, France
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | | | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Helen Lavretsky
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior
- David Geffen School of Medicine, University of California, Los Angeles
| | - Jonathan Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Jennifer Gatchel
- Department of Psychiatry, Massachusetts General Hospital, Boston
- McLean Hospital, Belmont, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Psychiatry, Baylor College of Medicine, Houston, Texas
- Michael E. DeBakey VA Medical Center, Houston, Texas
| | - Sandra Swantek
- American Association for Geriatric Psychiatry, Brentwood, Tennessee
| | - Paul Newhouse
- Vanderbilt University, Nashville, Tennessee
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University Medical Center, Nashville, Tennessee
- VA-TVHS Geriatric Research Education and Clinical Center, Nashville, Tennessee
| | - Howard H Feldman
- Department of Neurosciences, University of California, San Diego, La Jolla
- Shiley-Marcos Alzheimer's Disease Research Center, University of California, San Diego, La Jolla
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
- Memory Clinic, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
16
|
Moss DE, Perez RG. The phospho-tau cascade, basal forebrain neurodegeneration, and dementia in Alzheimer's disease: Anti-neurodegenerative benefits of acetylcholinesterase inhibitors. J Alzheimers Dis 2024; 102:617-626. [PMID: 39533696 DOI: 10.1177/13872877241289602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
A conundrum in Alzheimer's disease (AD) is why the long-term use of acetylcholinesterase (AChE) inhibitors, intended for treatment of dementia, results in slowing neurodegeneration in the cholinergic basal forebrain, hippocampus, and cortex. The phospho-tau cascade hypothesis presented here attempts to answer that question by unifying three hallmark features of AD into a specific sequence of events. It is proposed that the hyperphosphorylation of tau protein leads to the AD-associated deficit of nerve growth factor (NGF), then to atrophy of the cholinergic basal forebrain and dementia. Because the release of pro-nerve growth factor (pro-NGF) is activity-dependent and is controlled by basal forebrain projections to the hippocampus and cortex, our hypothesis is that AChE inhibitors act by increasing acetylcholine-dependent pro-NGF release and, thus, augmenting the availability of mature NGF and improving basal forebrain survival. If correct, improved central nervous system-selective AChE inhibitor therapy started prophylactically, before AD-associated basal forebrain atrophy and cognitive impairment onset, has the potential to delay not only the onset of dementia but also its rate of advancement. The phospho-tau hypothesis thus suggests that preventing hyperphosphorylation of tau protein, early should be a high priority as a strategy to help reduce dementia and its associated widespread social and economic suffering.
Collapse
Affiliation(s)
- Donald E Moss
- Professor Emeritus, University of Texas at El Paso, El Paso, TX, USA
| | | |
Collapse
|
17
|
Sola Fraca D, Sánchez Garrigós E, de Francisco Moure J, Marín Gonzalez B, Badiola Díez JJ, Acín Tresaco C. Sleep disturbance in clinical and preclinical scrapie-infected sheep measured by polysomnography. Vet Q 2024; 44:1-9. [PMID: 38698657 PMCID: PMC11073408 DOI: 10.1080/01652176.2024.2349674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 04/25/2024] [Indexed: 05/05/2024] Open
Abstract
Neurodegenerative diseases are characterised by neuronal loss and abnormal deposition of pathological proteins in the nervous system. Among the most common neurodegenerative diseases are Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease and transmissible spongiform encephalopathies (TSEs). Sleep and circadian rhythm disturbances are one of the most common symptoms in patients with neurodegenerative diseases. Currently, one of the main objectives in the study of TSEs is to try to establish an early diagnosis, as clinical signs do not appear until the damage to the central nervous system is very advanced, which prevents any therapeutic approach. In this paper, we provide the first description of sleep disturbance caused by classical scrapie in clinical and preclinical sheep using polysomnography compared to healthy controls. Fifteen sheep classified into three groups, clinical, preclinical and negative control, were analysed. The results show a decrease in total sleep time as the disease progresses, with significant changes between control, clinical and pre-clinical animals. The results also show an increase in sleep fragmentation in clinical animals compared to preclinical and control animals. In addition, sheep with clinical scrapie show a total loss of Rapid Eye Movement sleep (REM) and alterations in Non Rapid Eyes Movement sleep (NREM) compared to control sheep, demonstrating more shallow sleep. Although further research is needed, these results suggest that prion diseases also produce sleep disturbances in animals and that polysomnography could be a diagnostic tool of interest in clinical and preclinical cases of prion diseases.
Collapse
Affiliation(s)
- Diego Sola Fraca
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, Spain
| | | | | | - Belén Marín Gonzalez
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, Spain
| | - Juan José Badiola Díez
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, Spain
| | - Cristina Acín Tresaco
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, Spain
| |
Collapse
|
18
|
Brusini L, Dolci G, Pini L, Cruciani F, Pizzagalli F, Provero P, Menegaz G, Boscolo Galazzo I. Morphometric Similarity Patterning of Amyloid- β and Tau Proteins Correlates with Transcriptomics in the Alzheimer's Disease Continuum. Int J Mol Sci 2024; 25:12871. [PMID: 39684582 DOI: 10.3390/ijms252312871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Bridging the gap between cortical morphometric remodeling and gene expression can help to clarify the effects of the selective brain accumulation of Amyloid-β (Aβ) and tau proteins occurring in the Alzheimer's disease (AD). To this aim, we derived morphometric similarity (MS) networks from 126 Aβ- and tau-positive (Aβ+/tau+) and 172 Aβ-/tau- subjects, and we investigated the association between group-wise regional MS differences and transcriptional correlates thanks to an imaging transcriptomics approach grounded in the Allen Human Brain Atlas (AHBA). The expressed gene with the highest correlation with MS alterations was BCHE, a gene related to Aβ homeostasis. In addition, notably, among the most promising results derived from the enrichment analysis, we found the immune response to be a biological process and astrocytes, microglia, and oligodendrocyte precursors for the cell types. In summary, by relating cortical MS and AHBA-derived transcriptomics, we were able to retrieve findings suggesting the biological mechanisms underlying the Aβ- and tau- induced cortical MS alterations in the AD continuum.
Collapse
Affiliation(s)
- Lorenza Brusini
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy
| | - Giorgio Dolci
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy
- Department of Computer Science, University of Verona, 37134 Verona, Italy
| | - Lorenzo Pini
- Department of Neuroscience, University of Padova, 35121 Padova, Italy
| | - Federica Cruciani
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy
- Istituto Fondazione Oncologia Molecolare Ente del Terzo Settore (IFOM ETS)-The Associazione Italiana per la Ricerca sul Cancro (AIRC) Institute of Molecular Oncology, 20139 Milano, Italy
| | - Fabrizio Pizzagalli
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, 10126 Turin, Italy
| | - Paolo Provero
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, 10126 Turin, Italy
| | - Gloria Menegaz
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy
| | - Ilaria Boscolo Galazzo
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy
| |
Collapse
|
19
|
Ou CM, Xue WW, Liu D, Ma L, Xie HT, Ning K. Stem cell therapy in Alzheimer's disease: current status and perspectives. Front Neurosci 2024; 18:1440334. [PMID: 39640295 PMCID: PMC11618239 DOI: 10.3389/fnins.2024.1440334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
An incurable neurogenerative illness, Alzheimer's disease, is the cause of most global health, medical, and social disasters. The two main symptoms are cognitive impairment and neuronal loss. Current medications that target tau protein tangles and Aβ plaques are not very effective because they only slow the symptoms of AD and do not repair damaged cells. Stem cell-based treatments, however, present an alternative strategy in the treatment of AD. They have the capacity to divide into specialized adult cells, have self-renewal abilities, and multiplication. Stem cells can now be employed as a donor source for cell therapy due to developments in stem cell technology. This review covers preclinical and clinical updates on studies based on targeting the tau protein tangles and Aβ plaque, as well as four types of stem cells employed in AD treatment. The review also outlines the two basic pathologic aspects, tau protein tangles and Aβ plaques, of AD.
Collapse
Affiliation(s)
- Chu-Min Ou
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
| | - Wei-Wei Xue
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
- School of Biological Engineering, Dalian Polytechnic University, Dalian, China
| | - Dong Liu
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
| | - Liya Ma
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
| | - Hai-Tao Xie
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
| | - Ke Ning
- Guangdong Celconta Biotechnology Co., Ltd., Dongguan, Guangdong, China
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
20
|
Frisoni GB. Pathophysiology, diagnosis and care of Alzheimer's disease are coming together. J Neurol Neurosurg Psychiatry 2024:jnnp-2024-333864. [PMID: 39532520 DOI: 10.1136/jnnp-2024-333864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Giovanni B Frisoni
- Memory center, Geneva University and University Hospitals, Genève, Switzerland
| |
Collapse
|
21
|
Villain N, Michalon R. What is Alzheimer's disease? An analysis of nosological perspectives from the 20th and 21st centuries. Eur J Neurol 2024; 31:e16302. [PMID: 38618742 PMCID: PMC11464395 DOI: 10.1111/ene.16302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Recent US proposals suggest defining Alzheimer's disease (AD) based on β-amyloidosis alone. This sparked debates that echoed historical ones about the significance of brain lesions and clinical phenotype. METHODS This review covers debates on AD nosology through three key periods: AD's discovery in German-speaking countries in the early 20th century, its redefinition in Anglo-Saxon countries in the 1960s-1980s, and current debates on the biological or clinicobiological definitions of AD. Key players' opinions are focused on. RESULTS At the beginning of the 20th century, AD was defined as a clinicopathological entity. Debates arose around the pathological anchor, which included extended neurofibrillary tangles versus neuritic plaques (Alzheimer vs. Fischer) and its association with senile dementia (Kraepelin). In the 1960s-1980s, the debate shifted towards whether AD could be diagnosed using qualitative or quantitative neuropathological features and whether it was a unique process (Terry and Katzman) or had subtypes (Roth). The current definition proposed by the US Alzheimer's Association is based purely on biological β-amyloid abnormalities and represents a double break: from the historical clinicopathological definition of AD and from the historical emphasis on tau or combined tau and β-amyloid high levels of pathology. Conversely, the clinicobiological proposal of the International Working Group remains aligned with historical concepts of AD. CONCLUSIONS This historical perspective illustrates the unresolved questions surrounding AD pathogenesis, role of lesions, and the clinical phenotype, especially for sporadic cases. The intense nosological debates throughout the history of AD also illustrate the diversity of theoretical frameworks for defining disease in medicine.
Collapse
Affiliation(s)
- Nicolas Villain
- Sorbonne Université, INSERM U1127, CNRS 7225, Institut du Cerveau–ICMParisFrance
- Department of NeurologyInstitute of Memory and Alzheimer's Disease, AP‐HP Sorbonne Université, Pitié‐Salpêtrière HospitalParisFrance
| | - Robin Michalon
- École des Hautes Etudes en Sciences SocialesParisFrance
- CAK‐CRHST – Centre Alexandre Koyré – Centre de Recherche en Histoire des Sciences et des TechniquesParisFrance
| |
Collapse
|
22
|
Li J, Liu Y, Yin C, Zeng Y, Mei Y. Structural and functional remodeling of neural networks in β-amyloid driven hippocampal hyperactivity. Ageing Res Rev 2024; 101:102468. [PMID: 39218080 DOI: 10.1016/j.arr.2024.102468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Early detection of Alzheimer's disease (AD) is essential for improving the patients outcomes and advancing our understanding of disease, allowing for timely intervention and treatment. However, accurate biomarkers are still lacking. Recent evidence indicates that hippocampal hyperexcitability precedes the diagnosis of AD decades ago, can predict cognitive decline. Thus, could hippocampal hyperactivity be a robust biomarker for early-AD, and what drives hippocampal hyperactivity in early-AD? these critical questions remain to be answered. Increasing clinical and experimental studies suggest that early hippocampal activation is closely associated with longitudinal β-amyloid (Aβ) accumulation, Aβ aggregates, in turn, enhances hippocampal activity. Therefore, in this narrative review, we discuss the role of Aβ-induced altered intrinsic neuronal properties as well as structural and functional remodeling of glutamatergic, GABAergic, cholinergic, noradrenergic, serotonergic circuits in hippocampal hyperactivity. In addition, we analyze the available therapies and trials that can potentially be used clinically to attenuate hippocampal hyperexcitability in AD. Overall, the present review sheds lights on the mechanism behind Aβ-induced hippocampal hyperactivity, and highlights that hippocampal hyperactivity could be a robust biomarker and therapeutic target in prodromal AD.
Collapse
Affiliation(s)
- Jinquan Li
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yanjun Liu
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Chuhui Yin
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yan Zeng
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
23
|
Dorta S, Alexandre-Silva V, Popolin CP, de Sousa DB, Grigoli MM, Pelegrini LNDC, Manzine PR, Camins A, Marcello E, Endres K, Cominetti MR. ADAM10 isoforms: Optimizing usage of antibodies based on protein regulation, structural features, biological activity and clinical relevance to Alzheimer's disease. Ageing Res Rev 2024; 101:102464. [PMID: 39173916 DOI: 10.1016/j.arr.2024.102464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/21/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
A Disintegrin and Metalloproteinase 10 (ADAM10) is a crucial transmembrane protein involved in diverse cellular processes, including cell adhesion, migration, and proteolysis. ADAM10's ability to cleave over 100 substrates underscores its significance in physiological and pathological contexts, particularly in Alzheimer's disease (AD). This review comprehensively examines ADAM10's multifaceted roles, highlighting its critical function in the non-amyloidogenic processing of the amyloid precursor protein (APP), which mitigates amyloid beta (Aβ) production, a critical factor in AD development. We summarize the regulation of ADAM10 at multiple levels: transcriptional, translational, and post-translational, revealing the complexity and responsiveness of its expression to various cellular signals. A standardized nomenclature for ADAM10 isoforms is proposed to improve clarity and consistency in research, facilitating better comparison and replication of findings across studies. We address the challenges in detecting ADAM10 isoforms using antibodies, advocating for standardized detection protocols to resolve discrepancies in results from different biological matrices. By highlighting these issues, this review underscores the potential of ADAM10 as a biomarker for early diagnosis and a therapeutic target in AD. By consolidating current knowledge on ADAM10's regulation and function, we aim to provide insights that will guide future research and therapeutic strategies in the AD context.
Collapse
Affiliation(s)
- Sabrina Dorta
- Department of Gerontology, Federal University of São Carlos, São Carlos, SP, Brazil
| | | | | | | | | | | | | | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Marcia Regina Cominetti
- Department of Gerontology, Federal University of São Carlos, São Carlos, SP, Brazil; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
24
|
Kazemeini S, Nadeem-Tariq A, Shih R, Rafanan J, Ghani N, Vida TA. From Plaques to Pathways in Alzheimer's Disease: The Mitochondrial-Neurovascular-Metabolic Hypothesis. Int J Mol Sci 2024; 25:11720. [PMID: 39519272 PMCID: PMC11546801 DOI: 10.3390/ijms252111720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) presents a public health challenge due to its progressive neurodegeneration, cognitive decline, and memory loss. The amyloid cascade hypothesis, which postulates that the accumulation of amyloid-beta (Aβ) peptides initiates a cascade leading to AD, has dominated research and therapeutic strategies. The failure of recent Aβ-targeted therapies to yield conclusive benefits necessitates further exploration of AD pathology. This review proposes the Mitochondrial-Neurovascular-Metabolic (MNM) hypothesis, which integrates mitochondrial dysfunction, impaired neurovascular regulation, and systemic metabolic disturbances as interrelated contributors to AD pathogenesis. Mitochondrial dysfunction, a hallmark of AD, leads to oxidative stress and bioenergetic failure. Concurrently, the breakdown of the blood-brain barrier (BBB) and impaired cerebral blood flow, which characterize neurovascular dysregulation, accelerate neurodegeneration. Metabolic disturbances such as glucose hypometabolism and insulin resistance further impair neuronal function and survival. This hypothesis highlights the interconnectedness of these pathways and suggests that therapeutic strategies targeting mitochondrial health, neurovascular integrity, and metabolic regulation may offer more effective interventions. The MNM hypothesis addresses these multifaceted aspects of AD, providing a comprehensive framework for understanding disease progression and developing novel therapeutic approaches. This approach paves the way for developing innovative therapeutic strategies that could significantly improve outcomes for millions affected worldwide.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (S.K.); (A.N.-T.); (R.S.); (J.R.); (N.G.)
| |
Collapse
|
25
|
Wang YR, Zeng XQ, Wang J, Fowler CJ, Li QX, Bu XL, Doecke J, Maruff P, Martins RN, Rowe CC, Masters CL, Wang YJ, Liu YH. Autoantibodies to BACE1 promote Aβ accumulation and neurodegeneration in Alzheimer's disease. Acta Neuropathol 2024; 148:57. [PMID: 39448400 DOI: 10.1007/s00401-024-02814-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/14/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024]
Abstract
The profile of autoantibodies is dysregulated in patients with Alzheimer's disease (AD). Autoantibodies to beta-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) are present in human blood. This study aims to investigate the clinical relevance and pathophysiological roles of autoantibodies to BACE1 in AD. Clinical investigations were conducted in two independent cohorts, the Chongqing cohort, and the Australian Imaging, Biomarkers, and Lifestyle (AIBL) cohort. The Chongqing cohort included 55 AD patients, 28 patients with non-AD dementia, and 70 cognitively normal subjects (CN). The AIBL cohort included 162 Aβ-PET- CN, 169 Aβ-PET+ cognitively normal subjects (preclinical AD), and 31 Aβ-PET+ cognitively impaired subjects (Clinical AD). Plasma autoantibodies to BACE1 were determined by one-site Elisa. The associations of plasma autoantibodies to BACE1 with brain Aβ load and cognitive trajectory were investigated. The effects of autoantibodies to BACE1 on AD-type pathologies and underlying mechanisms were investigated in APP/PS1 mice and SH/APPswe/PS1wt cell lines. In the Chongqing cohort, plasma autoantibodies to BACE1 were higher in AD patients, in comparison with CN and non-AD dementia patients. In the AIBL cohort, plasma autoantibodies to BACE1 were highest in clinical AD patients, followed by preclinical AD and CN subjects. Higher autoantibodies to BACE1 were associated with an increased incidence of brain amyloid positivity conversion during follow-up. Autoantibodies to BACE1 exacerbated brain amyloid deposition and subsequent AD-type pathologies, including Tau hyperphosphorylation, neuroinflammation, and neurodegeneration in APP/PS1 mice. Autoantibodies to BACE1 increased Aβ production by promoting BACE1 expression through inhibiting PPARγ signaling. These findings suggest that autoantibodies to BACE1 are pathogenic in AD and the upregulation of these autoantibodies may promote the development of the disease. This study offers new insights into the mechanism of AD from an autoimmune perspective.
Collapse
Affiliation(s)
- Ye-Ran Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Aging and Brain Disease, Chongqing, China
- Centre of Health Management, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiao-Qin Zeng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Aging and Brain Disease, Chongqing, China
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Aging and Brain Disease, Chongqing, China
| | | | - Qiao-Xin Li
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Xian-Le Bu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Key Laboratory of Aging and Brain Disease, Chongqing, China
| | - James Doecke
- The Australian E-Health Research Centre, CSIRO, Herston, QLD, Australia
| | - Paul Maruff
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
- CogState, Melbourne, VIC, Australia
| | - Ralph N Martins
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
- Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Key Laboratory of Aging and Brain Disease, Chongqing, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Yu-Hui Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.
- Key Laboratory of Aging and Brain Disease, Chongqing, China.
| |
Collapse
|
26
|
Wen J, Yang Z, Nasrallah IM, Cui Y, Erus G, Srinivasan D, Abdulkadir A, Mamourian E, Hwang G, Singh A, Bergman M, Bao J, Varol E, Zhou Z, Boquet-Pujadas A, Chen J, Toga AW, Saykin AJ, Hohman TJ, Thompson PM, Villeneuve S, Gollub R, Sotiras A, Wittfeld K, Grabe HJ, Tosun D, Bilgel M, An Y, Marcus DS, LaMontagne P, Benzinger TL, Heckbert SR, Austin TR, Launer LJ, Espeland M, Masters CL, Maruff P, Fripp J, Johnson SC, Morris JC, Albert MS, Bryan RN, Resnick SM, Ferrucci L, Fan Y, Habes M, Wolk D, Shen L, Shou H, Davatzikos C. Genetic and clinical correlates of two neuroanatomical AI dimensions in the Alzheimer's disease continuum. Transl Psychiatry 2024; 14:420. [PMID: 39368996 PMCID: PMC11455841 DOI: 10.1038/s41398-024-03121-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024] Open
Abstract
Alzheimer's disease (AD) is associated with heterogeneous atrophy patterns. We employed a semi-supervised representation learning technique known as Surreal-GAN, through which we identified two latent dimensional representations of brain atrophy in symptomatic mild cognitive impairment (MCI) and AD patients: the "diffuse-AD" (R1) dimension shows widespread brain atrophy, and the "MTL-AD" (R2) dimension displays focal medial temporal lobe (MTL) atrophy. Critically, only R2 was associated with widely known sporadic AD genetic risk factors (e.g., APOE ε4) in MCI and AD patients at baseline. We then independently detected the presence of the two dimensions in the early stages by deploying the trained model in the general population and two cognitively unimpaired cohorts of asymptomatic participants. In the general population, genome-wide association studies found 77 genes unrelated to APOE differentially associated with R1 and R2. Functional analyses revealed that these genes were overrepresented in differentially expressed gene sets in organs beyond the brain (R1 and R2), including the heart (R1) and the pituitary gland, muscle, and kidney (R2). These genes were enriched in biological pathways implicated in dendritic cells (R2), macrophage functions (R1), and cancer (R1 and R2). Several of them were "druggable genes" for cancer (R1), inflammation (R1), cardiovascular diseases (R1), and diseases of the nervous system (R2). The longitudinal progression showed that APOE ε4, amyloid, and tau were associated with R2 at early asymptomatic stages, but this longitudinal association occurs only at late symptomatic stages in R1. Our findings deepen our understanding of the multifaceted pathogenesis of AD beyond the brain. In early asymptomatic stages, the two dimensions are associated with diverse pathological mechanisms, including cardiovascular diseases, inflammation, and hormonal dysfunction-driven by genes different from APOE-which may collectively contribute to the early pathogenesis of AD. All results are publicly available at https://labs-laboratory.com/medicine/ .
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA.
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Research Lab in Neuroimaging of the Department of Clinical Neurosciences at Lausanne University Hospital, Lausanne, Switzerland
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashish Singh
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Bergman
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erdem Varol
- Department of Statistics, Center for Theoretical Neuroscience, Zuckerman Institute, Columbia University, New York, NY, USA
| | - Zhen Zhou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aleix Boquet-Pujadas
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA
| | - Jiong Chen
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of NeuroImaging, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Andrew J Saykin
- Radiology and Imaging Sciences, Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research Center and the Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt Genetics Institute, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, McGill University, Montréal, QC, Canada
| | - Randy Gollub
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute for Informatics, Washington University School of Medicine, St. Louis, MO, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R Austin
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Lenore J Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Mark Espeland
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, QLD, Australia
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, 21225, USA
| | - Yong Fan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - David Wolk
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology and Penn Memory Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Min JH, Sarlus H, Harris RA. MAD-microbial (origin of) Alzheimer's disease hypothesis: from infection and the antimicrobial response to disruption of key copper-based systems. Front Neurosci 2024; 18:1467333. [PMID: 39416952 PMCID: PMC11480022 DOI: 10.3389/fnins.2024.1467333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Microbes have been suspected to cause Alzheimer's disease since at least 1908, but this has generally remained unpopular in comparison to the amyloid hypothesis and the dominance of Aβ and Tau. However, evidence has been accumulating to suggest that these earlier theories are but a manifestation of a common cause that can trigger and interact with all the major molecular players recognized in AD. Aβ, Tau and ApoE, in particular appear to be molecules with normal homeostatic functions but also with alternative antimicrobial functions. Their alternative functions confer the non-immune specialized neuron with some innate intracellular defenses that appear to be re-appropriated from their normal functions in times of need. Indeed, signs of infection of the neurons by biofilm-forming microbial colonies, in synergy with herpes viruses, are evident from the clinical and preclinical studies we discuss. Furthermore, we attempt to provide a mechanistic understanding of the AD landscape by discussing the antimicrobial effect of Aβ, Tau and ApoE and Lactoferrin in AD, and a possible mechanistic link with deficiency of vital copper-based systems. In particular, we focus on mitochondrial oxidative respiration via complex 4 and ceruloplasmin for iron homeostasis, and how this is similar and possibly central to neurodegenerative diseases in general. In the case of AD, we provide evidence for the microbial Alzheimer's disease (MAD) theory, namely that AD could in fact be caused by a long-term microbial exposure or even long-term infection of the neurons themselves that results in a costly prolonged antimicrobial response that disrupts copper-based systems that govern neurotransmission, iron homeostasis and respiration. Finally, we discuss potential treatment modalities based on this holistic understanding of AD that incorporates the many separate and seemingly conflicting theories. If the MAD theory is correct, then the reduction of microbial exposure through use of broad antimicrobial and anti-inflammatory treatments could potentially alleviate AD although this requires further clinical investigation.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital at Solna, Stockholm, Sweden
| | | | | |
Collapse
|
28
|
McGlinchey E, Duran-Aniotz C, Akinyemi R, Arshad F, Zimmer ER, Cho H, Adewale BA, Ibanez A. Biomarkers of neurodegeneration across the Global South. THE LANCET. HEALTHY LONGEVITY 2024; 5:100616. [PMID: 39369726 PMCID: PMC11540104 DOI: 10.1016/s2666-7568(24)00132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 10/08/2024] Open
Abstract
Research on neurodegenerative diseases has predominantly focused on high-income countries in the Global North. This Series paper describes the state of biomarker evidence for neurodegeneration in the Global South, including Latin America, Africa, and countries in south, east, and southeast Asia. Latin America shows growth in fluid biomarker and neuroimaging research, with notable advancements in genetics. Research in Africa focuses on genetics and cognition but there is a paucity of data on fluid and neuroimaging biomarkers. South and east Asia, particularly India and China, has achieved substantial progress in plasma, neuroimaging, and genetic studies. However, all three regions face several challenges in the form of a lack of harmonisation, insufficient funding, and few comparative studies both within the Global South, and between the Global North and Global South. Other barriers include scarce infrastructure, lack of knowledge centralisation, genetic and cultural diversity, sociocultural stigmas, and restricted access to tools such as PET scans. However, the diverse ethnic, genetic, economic, and cultural backgrounds in the Global South present unique opportunities for bidirectional learning, underscoring the need for global collaboration to enhance the understanding of dementia and brain health.
Collapse
Affiliation(s)
- Eimear McGlinchey
- Trinity College Dublin, Dublin, Ireland; Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| | - Claudia Duran-Aniotz
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibanez, Santiago de Chile, Chile
| | - Rufus Akinyemi
- Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria; Centre for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Faheem Arshad
- Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Eduardo R Zimmer
- Department of Pharmacology, Graduate Program in Biological Sciences: Pharmacology and Therapeutics (PPGFT) and Biochemistry (PPGBioq), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil; McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Hanna Cho
- Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Boluwatife Adeleye Adewale
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Agustin Ibanez
- Trinity College Dublin, Dublin, Ireland; Global Brain Health Institute, University of California San Francisco (UCSF), San Francisco, CA, USA; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland; Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibanez, Santiago de Chile, Chile.
| |
Collapse
|
29
|
Ibanez A, Slachevsky A. Environmental-genetic interactions in ageing and dementia across Latin America. Nat Rev Neurol 2024; 20:571-572. [PMID: 38997472 DOI: 10.1038/s41582-024-00998-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Affiliation(s)
- Agustin Ibanez
- Latin American Brain Health Institute, Universidad Adolfo Ibañez, Santiago, Chile.
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| | - Andrea Slachevsky
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
- Memory and Neuropsychiatric Center, Neurology Department, Hospital del Salvador & Faculty of Medicine, University of Chile, Santiago, Chile.
- Neuropsychology and Clinical Neuroscience Laboratory, Physiopathology Department, Institute of Biomedical Sciences, Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile.
- Neurology and Psychiatry Department, Clínica Alemana-University Desarrollo, Santiago, Chile.
| |
Collapse
|
30
|
Granzotto A, Vissel B, Sensi SL. Lost in translation: Inconvenient truths on the utility of mouse models in Alzheimer's disease research. eLife 2024; 13:e90633. [PMID: 39329365 PMCID: PMC11434637 DOI: 10.7554/elife.90633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
The recent, controversial approval of antibody-based treatments for Alzheimer's disease (AD) is fueling a heated debate on the molecular determinants of this condition. The discussion should also incorporate a critical revision of the limitations of preclinical mouse models in advancing our understanding of AD. We critically discuss the limitations of animal models, stressing the need for careful consideration of how experiments are designed and results interpreted. We identify the shortcomings of AD models to recapitulate the complexity of the human disease. We dissect these issues at the quantitative, qualitative, temporal, and context-dependent levels. We argue that these models are based on the oversimplistic assumptions proposed by the amyloid cascade hypothesis (ACH) of AD and fail to account for the multifactorial nature of the condition. By shedding light on the constraints of current experimental tools, this review aims to foster the development and implementation of more clinically relevant tools. While we do not rule out a role for preclinical models, we call for alternative approaches to be explored and, most importantly, for a re-evaluation of the ACH.
Collapse
Affiliation(s)
- Alberto Granzotto
- Center for Advanced Studies and Technology – CAST, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d’Annunzio of Chieti-PescaraChietiItaly
| | - Bryce Vissel
- St Vincent’s Hospital Centre for Applied Medical Research, St Vincent’s HospitalDarlinghurstAustralia
- School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyAustralia
| | - Stefano L Sensi
- Center for Advanced Studies and Technology – CAST, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Institute for Advanced Biomedical Technologies – ITAB, University G. d’Annunzio of Chieti-PescaraChietiItaly
- Institute of Neurology, SS Annunziata University Hospital, University G. d’Annunzio of Chieti-PescaraChietiItaly
| |
Collapse
|
31
|
Zhang X, Lu Q, Meng X, Kim JY. Protocol to induce neurodegeneration in a local area of the mouse brain by stereotaxic injection. STAR Protoc 2024; 5:103243. [PMID: 39096497 PMCID: PMC11342772 DOI: 10.1016/j.xpro.2024.103243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 08/05/2024] Open
Abstract
In vivo models of brain pathology are crucial for studying neurological diseases. Here, we present a protocol to induce a pathological condition in a mouse brain area by local injection of neurotoxic stimulus. We describe steps for preparing reagents, stereotaxic injection procedures to induce neurodegeneration in the hippocampus, and preparation of brain sections to examine the induced model. This protocol is useful for studying how local pathology affects other brain areas and neighbor cells and its functional consequences in behavior. For complete details on the use and execution of this protocol, please refer to Zhang et al.1.
Collapse
Affiliation(s)
- Xuebing Zhang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Qingqing Lu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Xingqi Meng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Jin Young Kim
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
32
|
Arshavsky YI. Autoimmune hypothesis of Alzheimer's disease: unanswered question. J Neurophysiol 2024; 132:929-942. [PMID: 39163023 DOI: 10.1152/jn.00259.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alzheimer's disease (AD) was described more than a century ago. However, there are still no effective approaches to its treatment, which may suggest that the search for the cure is not being conducted in the most productive direction. AD begins as selective impairments of declarative memory with no deficits in other cognitive functions. Therefore, understanding of the AD pathogenesis has to include the understanding of this selectivity. Currently, the main efforts aimed at prevention and treatment of AD are based on the dominating hypothesis for the AD pathogenesis: the amyloid hypothesis. But this hypothesis does not explain selective memory impairments since β-amyloid accumulates extracellularly and should be toxic to all types of cerebral neurons, not only to "memory engram neurons." To explain selective memory impairment, I propose the autoimmune hypothesis of AD, based on the analysis of risk factors for AD and molecular mechanisms of memory formation. Memory formation is associated with epigenetic modifications of chromatin in memory engram neurons and, therefore, might be accompanied by the expression of memory-specific proteins recognized by the adaptive immune system as "non-self" antigens. Normally, the brain is protected by the blood-brain barrier (BBB). All risk factors for AD provoke BBB disruptions, possibly leading to an autoimmune reaction against memory engram neurons. This reaction would make them selectively sensitive to tauopathy. If this hypothesis is confirmed, the strategies for AD prevention and treatment would be radically changed.
Collapse
Affiliation(s)
- Yuri I Arshavsky
- BioCircuits Institute, University of California, San Diego, La Jolla, California, United States
| |
Collapse
|
33
|
Daly T. A philosophy of science approach to the amyloid hypothesis of Alzheimer's disease. Eur J Neurosci 2024; 60:4707-4722. [PMID: 39119857 DOI: 10.1111/ejn.16500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/04/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Disputes about the scientific validity of the amyloid-β hypothesis of Alzheimer's disease have been held since the early 1990s, with little constructive progress made between opposing sides despite recent therapeutic progress. Here, I argue that philosophy of science can improve the chance of constructive debate by giving researchers technical language to describe and assess scientific progress. To do so, I interpret the amyloid hypothesis using a modified version of the research programme concept from philosopher of science Imre Lakatos. I first outline the amyloid-β hypothesis and study critiques of its central place in Alzheimer's research. Then, I draw on the complexity of amyloid-β and Alzheimer's research to discuss the limits of using concepts from popular philosophers of science Karl Popper or Thomas Kuhn, before finally arguing that an adaptation of the research programme concept can foster constructive debates about the science of Alzheimer's and within it. I will argue that the amyloid-β hypothesis has contributed to significant progress in the Alzheimer's field based on what Lakatos called the "positive heuristic" (motivating the programme to test its predictions) and the "negative heuristic" (protecting the programme from refutation). I consider the amyloid research agenda to be progressive despite the fact that its claims about disease aetiology could be wrong.
Collapse
Affiliation(s)
- Timothy Daly
- Bioethics Program, FLACSO Argentina, Buenos Aires, Argentina
- Science Norms Democracy UMR 8011, Sorbonne Université, Paris, France
| |
Collapse
|
34
|
Huang Q, Wu W, Wen Y, Lu S, Zhao C. Potential therapeutic natural compounds for the treatment of Alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155822. [PMID: 38909512 DOI: 10.1016/j.phymed.2024.155822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a complicated neurodegenerative disease with cognitive impairment occurring in the older people, in which extracellular accumulation of β-amyloid and intracellular aggregation of hyperphosphorylated tau are regarded as the prevailing theories. However, the exact AD mechanism has not been determined. Moreover, there is no effective treatment available in phase III trials to eradicate AD, which is imperative to explore novel treatments. PURPOSE A number of up-to-date pre-clinical studies on cognitive impairment is beneficial to clarify the pathology of AD. This review recapitulates several advances in AD pathobiology and discusses the neuroprotective effects of natural compounds, such as phenolic compounds, natural polysaccharides and oligosaccharides, peptide, and lipids, underscoring the therapeutic potential for AD. METHODS Electronic databases involving PubMed, Web of Science, and Google Scholar were searched up to October 2023. Articles were conducted using the keywords like Alzheimer's disease, pathogenic mechanisms, natural compounds, and neuroprotection. RESULT This review summarized several AD pathologies and the neuroprotective effects of natural compounds such as natural polysaccharides and oligosaccharides, peptide, and lipids. CONCLUSION We have discussed the pathogenic mechanisms of AD and the effect natural products on neurodegenerative diseases particularly in treating AD. Specifically, we investigated the molecular pathways and links between natural compounds and Alzheimer's disease such as through NF-κB, Nrf2, and mTOR pathway. Further investigation is necessary in exploring the bioactivity and effectiveness of natural compounds in clinical trials, which may provide a promising treatment for AD patients.
Collapse
Affiliation(s)
- Qihui Huang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, 36310 Vigo, Spain
| | - Weihao Wu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yuxi Wen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA)-CITEXVI, 36310 Vigo, Spain
| | - Suyue Lu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chao Zhao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
35
|
Hsiao PJ, Wu CD, Wong PY, Chung MC, Yang YW, Wu LY, Hsiao KY, Chung CJ. APOE-ε4 Alleles Modify the Decline of MMSE Scores Associated With Time-Dependent PM 2.5 Exposure: Findings From a Community-Based Longitudinal Cohort Study. Am J Geriatr Psychiatry 2024; 32:1080-1092. [PMID: 38228451 DOI: 10.1016/j.jagp.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 01/18/2024]
Abstract
OBJECTIVE Limited research has explored the long-term effect of reduced PM2.5 exposure on cognitive function. This study aimed to investigate the effects of time-dependent PM2.5 exposure and the interactions of PM2.5 and aging on declines in Mini-Mental State Examination (MMSE) scores, in carriers and non-carriers of the APOE-ε4 allele. METHODS Participants aged over 60 were recruited for this cohort study, undergoing MMSE tests twice from the Taiwan Biobank Program from 2008 to 2020. Participants with dementia or baseline MMSE scores <24 were excluded. Annual PM2.5 levels were estimated using a hybrid kriging/land use regression model with extreme gradient boosting, treated as a time-dependent variable. Generalized estimating equations were used to assess the impacts of repeated PM2.5 on MMSE decline, further stratified by the presence of APOE-ε4 alleles. RESULTS After follow-up, 290 participants out of the overall 7,000 community residents in the Biobank dataset demonstrated incidences of MMSE declines (<24), with an average MMSE score decline of 1.11 per year. Participants with ε4/ε4 alleles in the APOE gene had significantly 3.68-fold risks of MMSE decline. High levels of PM2.5 across all visits were significantly associated with worsening of scores on the overall MMSE. As annual levels of PM2.5 decreased over time, the impact of PM2.5 on MMSE decline also slowly diminished. CONCLUSION Long-term PM2.5 exposure may be associated with increased risk of MMSE decline, despite improvements in ambient PM2.5 levels over time. Validation of these results necessitates a large-scale prospective cohort study with more concise cognitive screening tools.
Collapse
Affiliation(s)
- Po-Jen Hsiao
- Department of Urology (P-JH), China Medical University and Hospital, Taichung, Taiwan
| | - Chih-Da Wu
- Department of Geomatics (C-DW), National Cheng Kung University, Tainan, Taiwan; National Institute of Environmental Health Sciences (C-DW), National Health Research Institutes, Miaoli, Taiwan; Innovation and Development Center of Sustainable Agriculture (C-DW), National Chung Hsing University, Taichung, Taiwan
| | - Pei-Yi Wong
- Department of Environmental and Occupational Health (P-YW), National Cheng Kung University, Tainan, Taiwan
| | - Mu-Chi Chung
- Division of Nephrology, Department of Medicine (M-CC), Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yu-Wan Yang
- Department of Neurology (Y-WY), China Medical University and Hospital, Taichung, Taiwan
| | - Laing-You Wu
- Department of Public Health (L-YW, C-JC), College of Public Health, China Medical University, Taichung, Taiwan
| | - Kai-Yu Hsiao
- Division of Thoracic Surgery (K-YH), Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Jung Chung
- Department of Public Health (L-YW, C-JC), College of Public Health, China Medical University, Taichung, Taiwan; Department of Medical Research (C-JC), China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
36
|
Koszła O, Sołek P. Misfolding and aggregation in neurodegenerative diseases: protein quality control machinery as potential therapeutic clearance pathways. Cell Commun Signal 2024; 22:421. [PMID: 39215343 PMCID: PMC11365204 DOI: 10.1186/s12964-024-01791-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
The primary challenge in today's world of neuroscience is the search for new therapeutic possibilities for neurodegenerative disease. Central to these disorders lies among other factors, the aberrant folding, aggregation, and accumulation of proteins, resulting in the formation of toxic entities that contribute to neuronal degeneration. This review concentrates on the key proteins such as β-amyloid (Aβ), tau, and α-synuclein, elucidating the intricate molecular events underlying their misfolding and aggregation. We critically evaluate the molecular mechanisms governing the elimination of misfolded proteins, shedding light on potential therapeutic strategies. We specifically examine pathways such as the endoplasmic reticulum (ER) and unfolded protein response (UPR), chaperones, chaperone-mediated autophagy (CMA), and the intersecting signaling of Keap1-Nrf2-ARE, along with autophagy connected through p62. Above all, we emphasize the significance of these pathways as protein quality control mechanisms, encompassing interventions targeting protein aggregation, regulation of post-translational modifications, and enhancement of molecular chaperones and clearance. Additionally, we focus on current therapeutic possibilities and new, multi-target approaches. In conclusion, this review systematically consolidates insights into emerging therapeutic strategies predicated on protein aggregates clearance.
Collapse
Affiliation(s)
- Oliwia Koszła
- Department of Biopharmacy, Medical University of Lublin, 4A Chodzki St., Lublin, 20-093, Poland.
| | - Przemysław Sołek
- Department of Biopharmacy, Medical University of Lublin, 4A Chodzki St., Lublin, 20-093, Poland
- Department of Biochemistry and Toxicology, University of Life Sciences, 13 Akademicka St, Lublin, 20-950, Poland
| |
Collapse
|
37
|
Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, Chen L. Recent advances in Alzheimer's disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduct Target Ther 2024; 9:211. [PMID: 39174535 PMCID: PMC11344989 DOI: 10.1038/s41392-024-01911-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) stands as the predominant form of dementia, presenting significant and escalating global challenges. Its etiology is intricate and diverse, stemming from a combination of factors such as aging, genetics, and environment. Our current understanding of AD pathologies involves various hypotheses, such as the cholinergic, amyloid, tau protein, inflammatory, oxidative stress, metal ion, glutamate excitotoxicity, microbiota-gut-brain axis, and abnormal autophagy. Nonetheless, unraveling the interplay among these pathological aspects and pinpointing the primary initiators of AD require further elucidation and validation. In the past decades, most clinical drugs have been discontinued due to limited effectiveness or adverse effects. Presently, available drugs primarily offer symptomatic relief and often accompanied by undesirable side effects. However, recent approvals of aducanumab (1) and lecanemab (2) by the Food and Drug Administration (FDA) present the potential in disrease-modifying effects. Nevertheless, the long-term efficacy and safety of these drugs need further validation. Consequently, the quest for safer and more effective AD drugs persists as a formidable and pressing task. This review discusses the current understanding of AD pathogenesis, advances in diagnostic biomarkers, the latest updates of clinical trials, and emerging technologies for AD drug development. We highlight recent progress in the discovery of selective inhibitors, dual-target inhibitors, allosteric modulators, covalent inhibitors, proteolysis-targeting chimeras (PROTACs), and protein-protein interaction (PPI) modulators. Our goal is to provide insights into the prospective development and clinical application of novel AD drugs.
Collapse
Affiliation(s)
- Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yinglu Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, TN, USA
| | - Yilin Xia
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxian Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
38
|
Giorgio J, Jonson C, Wang Y, Yokoyama JS, Wang J, Jagust W. Variable and interactive effects of Sex, APOE ε4 and TREM2 on the deposition of tau in entorhinal and neocortical regions. RESEARCH SQUARE 2024:rs.3.rs-4804430. [PMID: 39149503 PMCID: PMC11326369 DOI: 10.21203/rs.3.rs-4804430/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The canonical AD pathological cascade posits that the accumulation of amyloid beta ( Aβ ) is the initiating event, accelerating the accumulation of tau in the entorhinal cortex (EC), which subsequently spreads into the neocortex. Here in a sample of over 1300 participants with multimodal imaging and genetic information we queried how genetic variation affects these stages of the AD cascade. We observed that females and APOE- ε4 homozygotes are more susceptible to the effects of Aβ on the primary accumulation of tau, with greater EC tau for a given level of Aβ . Furthermore, we observed for individuals who have rare risk variants in Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) and/or APOE- ε4 homozygotes there was a greater spread of primary tau from the EC into the neocortex. These findings offer insights into the function of sex, APOE and microglia in AD progression, and have implications for determining personalised treatment with drugs targeting Aβ and tau.
Collapse
Affiliation(s)
- Joseph Giorgio
- Department of Neuroscience, University of California Berkeley, Berkeley, California, USA, 94720
- School of Psychological Sciences, College of Engineering, Science and the Environment, University of Newcastle, Newcastle, New South Wales, Australia, 2308
| | - Caroline Jonson
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD USA 20892
- DataTecnica LLC, Washington, DC, USA, 20037
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, USA, 94158
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA, 94158
| | - Yilin Wang
- Department of Statistics and Actuarial Science, The University of Iowa, Iowa City, IA, USA
| | - Jennifer S. Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA, 94158
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Jingshen Wang
- Division of Biostatistics, University of California Berkeley, Berkeley, California, USA, 94720
| | - William Jagust
- Department of Neuroscience, University of California Berkeley, Berkeley, California, USA, 94720
| | | | | |
Collapse
|
39
|
Meghdadi AH, Salat D, Hamilton J, Hong Y, Boeve BF, St Louis EK, Verma A, Berka C. EEG and ERP biosignatures of mild cognitive impairment for longitudinal monitoring of early cognitive decline in Alzheimer's disease. PLoS One 2024; 19:e0308137. [PMID: 39116138 PMCID: PMC11309464 DOI: 10.1371/journal.pone.0308137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Cognitive decline in Alzheimer's disease is associated with electroencephalographic (EEG) biosignatures even at early stages of mild cognitive impairment (MCI). The aim of this work is to provide a unified measure of cognitive decline by aggregating biosignatures from multiple EEG modalities and to evaluate repeatability of the composite measure at an individual level. These modalities included resting state EEG (eyes-closed) and two event-related potential (ERP) tasks on visual memory and attention. We compared individuals with MCI (n = 38) to age-matched healthy controls HC (n = 44). In resting state EEG, the MCI group exhibited higher power in Theta (3-7Hz) and lower power in Beta (13-20Hz) frequency bands. In both ERP tasks, the MCI group exhibited reduced ERP late positive potential (LPP), delayed ERP early component latency, slower reaction time, and decreased response accuracy. Cluster-based permutation analysis revealed significant clusters of difference between the MCI and HC groups in the frequency-channel and time-channel spaces. Cluster-based measures and performance measures (12 biosignatures in total) were selected as predictors of MCI. We trained a support vector machine (SVM) classifier achieving AUC = 0.89, accuracy = 77% in cross-validation using all data. Split-data validation resulted in (AUC = 0.87, accuracy = 76%) and (AUC = 0.75, accuracy = 70%) on testing data at baseline and follow-up visits, respectively. Classification scores at baseline and follow-up visits were correlated (r = 0.72, p<0.001, ICC = 0.84), supporting test-retest reliability of EEG biosignature. These results support the utility of EEG/ERP for prognostic testing, repeated assessments, and tracking potential treatment outcomes in the limited duration of clinical trials.
Collapse
Affiliation(s)
- Amir H. Meghdadi
- Advanced Brain Monitoring, Inc., Carlsbad, CA, United States of America
| | - David Salat
- Massachusetts General Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | | | - Yue Hong
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Bradley F. Boeve
- Departments of Neurology and Medicine, Division of Pulmonary and Critical Care Medicine, Center for Sleep Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, United States of America
| | - Erik K. St Louis
- Departments of Neurology and Medicine, Division of Pulmonary and Critical Care Medicine, Center for Sleep Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, United States of America
- Department of Clinical and Translational Research, Mayo Clinic Health System Southwest Wisconsin, La Crosse, WI, United States of America
| | - Ajay Verma
- Formation Venture Engineering, Boston, MA, United States of America
| | - Chris Berka
- Advanced Brain Monitoring, Inc., Carlsbad, CA, United States of America
| |
Collapse
|
40
|
Brusini L, Cruciani F, Dall’Aglio G, Zajac T, Boscolo Galazzo I, Zucchelli M, Menegaz G. XAI-Based Assessment of the AMURA Model for Detecting Amyloid-β and Tau Microstructural Signatures in Alzheimer's Disease. IEEE JOURNAL OF TRANSLATIONAL ENGINEERING IN HEALTH AND MEDICINE 2024; 12:569-579. [PMID: 39155922 PMCID: PMC11329216 DOI: 10.1109/jtehm.2024.3430035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 05/24/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024]
Abstract
Brain microstructural changes already occur in the earliest phases of Alzheimer's disease (AD) as evidenced in diffusion magnetic resonance imaging (dMRI) literature. This study investigates the potential of the novel dMRI Apparent Measures Using Reduced Acquisitions (AMURA) as imaging markers for capturing such tissue modifications.Tract-based spatial statistics (TBSS) and support vector machines (SVMs) based on different measures were exploited to distinguish between amyloid-beta/tau negative (A[Formula: see text]-/tau-) and A[Formula: see text]+/tau+ or A[Formula: see text]+/tau- subjects. Moreover, eXplainable Artificial Intelligence (XAI) was used to highlight the most influential features in the SVMs classifications and to validate the results by seeing the explanations' recurrence across different methods.TBSS analysis revealed significant differences between A[Formula: see text]-/tau- and other groups in line with the literature. The best SVM classification performance reached an accuracy of 0.73 by using advanced measures compared to more standard ones. Moreover, the explainability analysis suggested the results' stability and the central role of the cingulum to show early sign of AD.By relying on SVM classification and XAI interpretation of the outcomes, AMURA indices can be considered viable markers for amyloid and tau pathology. Clinical impact: This pre-clinical research revealed AMURA indices as viable imaging markers for timely AD diagnosis by acquiring clinically feasible dMR images, with advantages compared to more invasive methods employed nowadays.
Collapse
Affiliation(s)
- Lorenza Brusini
- Department of Engineering for Innovation MedicineUniversity of VeronaVerona37134Italy
| | - Federica Cruciani
- Department of Engineering for Innovation MedicineUniversity of VeronaVerona37134Italy
| | | | - Tommaso Zajac
- Department of Computer ScienceUniversity of VeronaVerona37134Italy
| | | | - Mauro Zucchelli
- Department of Research and Development Advanced ApplicationsOlea MedicalLa Ciotat13600France
| | - Gloria Menegaz
- Department of Engineering for Innovation MedicineUniversity of VeronaVerona37134Italy
| |
Collapse
|
41
|
Petralla S, Panayotova M, Franchina E, Fricker G, Puris E. Low-Density Lipoprotein Receptor-Related Protein 1 as a Potential Therapeutic Target in Alzheimer's Disease. Pharmaceutics 2024; 16:948. [PMID: 39065645 PMCID: PMC11279518 DOI: 10.3390/pharmaceutics16070948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease impacting the lives of millions of people worldwide. The formation of amyloid β (Aβ) plagues in the brain is the main pathological hallmark of AD. The Aβ deposits are formed due to the imbalance between the production and Aβ clearance in the brain and across the blood-brain barrier (BBB). In this respect, low-density lipoprotein receptor-related protein 1 (LRP1) plays a significant role by mediating both brain Aβ production and clearance. Due to its important role in AD pathogenesis, LRP1 is considered an attractive drug target for AD therapies. In the present review, we summarize the current knowledge about the role of LRP1 in AD pathogenesis as well as recent findings on changes in LRP1 expression and function in AD. Finally, we discuss the advances in utilizing LRP1 as a drug target for AD treatments as well as future perspectives on LRP1 research.
Collapse
Affiliation(s)
| | | | | | | | - Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany; (S.P.); (M.P.); (E.F.); (G.F.)
| |
Collapse
|
42
|
Liu Y, Xia X, Zheng M, Shi B. Bio-Nano Toolbox for Precision Alzheimer's Disease Gene Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314354. [PMID: 38778446 DOI: 10.1002/adma.202314354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's disease (AD) is the most burdensome aging-associated neurodegenerative disorder, and its treatment encounters numerous failures during drug development. Although there are newly approved in-market β-amyloid targeting antibody solutions, pathological heterogeneity among patient populations still challenges the treatment outcome. Emerging advances in gene therapies offer opportunities for more precise personalized medicine; while, major obstacles including the pathological heterogeneity among patient populations, the puzzled mechanism for druggable target development, and the precision delivery of functional therapeutic elements across the blood-brain barrier remain and limit the use of gene therapy for central neuronal diseases. Aiming for "precision delivery" challenges, nanomedicine provides versatile platforms that may overcome the targeted delivery challenges for AD gene therapy. In this perspective, to picture a toolbox for AD gene therapy strategy development, the most recent advances from benchtop to clinics are highlighted, possibly available gene therapy targets, tools, and delivery platforms are outlined, their challenges as well as rational design elements are addressed, and perspectives in this promising research field are discussed.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xue Xia
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| |
Collapse
|
43
|
Sasner M, Preuss C, Pandey RS, Uyar A, Garceau D, Kotredes KP, Williams H, Oblak AL, Lin PB, Perkins B, Soni D, Ingraham C, Lee‐Gosselin A, Lamb BT, Howell GR, Carter GW. In vivo validation of late-onset Alzheimer's disease genetic risk factors. Alzheimers Dement 2024; 20:4970-4984. [PMID: 38687251 PMCID: PMC11247676 DOI: 10.1002/alz.13840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Genome-wide association studies have identified over 70 genetic loci associated with late-onset Alzheimer's disease (LOAD), but few candidate polymorphisms have been functionally assessed for disease relevance and mechanism of action. METHODS Candidate genetic risk variants were informatically prioritized and individually engineered into a LOAD-sensitized mouse model that carries the AD risk variants APOE ε4/ε4 and Trem2*R47H. The potential disease relevance of each model was assessed by comparing brain transcriptomes measured with the Nanostring Mouse AD Panel at 4 and 12 months of age with human study cohorts. RESULTS We created new models for 11 coding and loss-of-function risk variants. Transcriptomic effects from multiple genetic variants recapitulated a variety of human gene expression patterns observed in LOAD study cohorts. Specific models matched to emerging molecular LOAD subtypes. DISCUSSION These results provide an initial functionalization of 11 candidate risk variants and identify potential preclinical models for testing targeted therapeutics. HIGHLIGHTS A novel approach to validate genetic risk factors for late-onset AD (LOAD) is presented. LOAD risk variants were knocked in to conserved mouse loci. Variant effects were assayed by transcriptional analysis. Risk variants in Abca7, Mthfr, Plcg2, and Sorl1 loci modeled molecular signatures of clinical disease. This approach should generate more translationally relevant animal models.
Collapse
Affiliation(s)
| | | | - Ravi S. Pandey
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Asli Uyar
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | | | | | | | - Adrian L. Oblak
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Peter Bor‐Chian Lin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bridget Perkins
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Disha Soni
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Cindy Ingraham
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Audrey Lee‐Gosselin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bruce T. Lamb
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | | | - Gregory W. Carter
- The Jackson LaboratoryBar HarborMaineUSA
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| |
Collapse
|
44
|
Guo W, Liu Y, Han Y, Tang H, Fan X, Wang C, Chen PR. Amplifiable protein identification via residue-resolved barcoding and composition code counting. Natl Sci Rev 2024; 11:nwae183. [PMID: 39055168 PMCID: PMC11272068 DOI: 10.1093/nsr/nwae183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 07/27/2024] Open
Abstract
Ultrasensitive protein identification is of paramount importance in basic research and clinical diagnostics but remains extremely challenging. A key bottleneck in preventing single-molecule protein sequencing is that, unlike the revolutionary nucleic acid sequencing methods that rely on the polymerase chain reaction (PCR) to amplify DNA and RNA molecules, protein molecules cannot be directly amplified. Decoding the proteins via amplification of certain fingerprints rather than the intact protein sequence thus represents an appealing alternative choice to address this formidable challenge. Herein, we report a proof-of-concept method that relies on residue-resolved DNA barcoding and composition code counting for amplifiable protein fingerprinting (AmproCode). In AmproCode, selective types of residues on peptides or proteins are chemically labeled with a DNA barcode, which can be amplified and quantified via quantitative PCR. The operation generates a relative ratio as the residue-resolved 'composition code' for each target protein that can be utilized as the fingerprint to determine its identity from the proteome database. We developed a database searching algorithm and applied it to assess the coverage of the whole proteome and secretome via computational simulations, proving the theoretical feasibility of AmproCode. We then designed the residue-specific DNA barcoding and amplification workflow, and identified different synthetic model peptides found in the secretome at as low as the fmol/L level for demonstration. These results build the foundation for an unprecedented amplifiable protein fingerprinting method. We believe that, in the future, AmproCode could ultimately realize single-molecule amplifiable identification of trace complex samples without further purification, and it may open a new avenue in the development of next-generation protein sequencing techniques.
Collapse
Affiliation(s)
- Weiming Guo
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yuan Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yu Han
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Huan Tang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Xinyuan Fan
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chu Wang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Peng R Chen
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
45
|
Stockinger F, Poc P, Möhwald A, Karch S, Häfner S, Alzheimer C, Sandoz G, Huth T, Broichhagen J. Multicolor, Cell-Impermeable, and High Affinity BACE1 Inhibitor Probes Enable Superior Endogenous Staining and Imaging of Single Molecules. J Med Chem 2024; 67:10152-10167. [PMID: 38842406 PMCID: PMC11215771 DOI: 10.1021/acs.jmedchem.4c00339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/10/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
The prevailing but not undisputed amyloid cascade hypothesis places the β-site of APP cleaving enzyme 1 (BACE1) center stage in Alzheimer's Disease pathogenesis. Here, we investigated functional properties of BACE1 with novel tag- and antibody-free labeling tools, which are conjugates of the BACE1-inhibitor IV (also referred to as C3) linked to different impermeable Alexa Fluor dyes. We show that these fluorescent small molecules bind specifically to BACE1, with a 1:1 labeling stoichiometry at their orthosteric site. This is a crucial property especially for single-molecule and super-resolution microscopy approaches, allowing characterization of the dyes' labeling capabilities in overexpressing cell systems and in native neuronal tissue. With multiple colors at hand, we evaluated BACE1-multimerization by Förster resonance energy transfer (FRET) acceptor-photobleaching and single-particle imaging of native BACE1. In summary, our novel fluorescent inhibitors, termed Alexa-C3, offer unprecedented insights into protein-protein interactions and diffusion behavior of BACE1 down to the single molecule level.
Collapse
Affiliation(s)
- Florian Stockinger
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Pascal Poc
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Heidelberg 69120, Germany
- Leibniz-Forschungsinstitut
für Molekulare Pharmakologie, Berlin 13125, Germany
| | - Alexander Möhwald
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Sandra Karch
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Stephanie Häfner
- Université
Côte d’Azur, CNRS, INSERM,
iBV, Nice 06108, Cedex 2, France
- Laboratories
of Excellence, Ion Channel Science and Therapeutics, Nice 06108, Cedex 2, France
| | - Christian Alzheimer
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Guillaume Sandoz
- Université
Côte d’Azur, CNRS, INSERM,
iBV, Nice 06108, Cedex 2, France
- Laboratories
of Excellence, Ion Channel Science and Therapeutics, Nice 06108, Cedex 2, France
| | - Tobias Huth
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Johannes Broichhagen
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Heidelberg 69120, Germany
- Leibniz-Forschungsinstitut
für Molekulare Pharmakologie, Berlin 13125, Germany
| |
Collapse
|
46
|
Calis S, Gevaert K. The role of Nα-terminal acetylation in protein conformation. FEBS J 2024. [PMID: 38923676 DOI: 10.1111/febs.17209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Especially in higher eukaryotes, the N termini of proteins are subject to enzymatic modifications, with the acetylation of the alpha-amino group of nascent polypeptides being a prominent one. In recent years, the specificities and substrates of the enzymes responsible for this modification, the Nα-terminal acetyltransferases, have been mapped in several proteomic studies. Aberrant expression of, and mutations in these enzymes were found to be associated with several human diseases, explaining the growing interest in protein Nα-terminal acetylation. With some enzymes, such as the Nα-terminal acetyltransferase A complex having thousands of possible substrates, researchers are now trying to decipher the functional outcome of Nα-terminal protein acetylation. In this review, we zoom in on one possible functional consequence of Nα-terminal protein acetylation; its effect on protein folding. Using selected examples of proteins associated with human diseases such as alpha-synuclein and huntingtin, here, we discuss the sometimes contradictory findings of the effects of Nα-terminal protein acetylation on protein (mis)folding and aggregation.
Collapse
Affiliation(s)
- Sam Calis
- VIB Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Belgium
| | - Kris Gevaert
- VIB Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Belgium
| |
Collapse
|
47
|
Enzlein T, Lashley T, Sammour DA, Hopf C, Chávez-Gutiérrez L. Integrative Single-Plaque Analysis Reveals Signature Aβ and Lipid Profiles in the Alzheimer's Brain. Anal Chem 2024; 96:9799-9807. [PMID: 38830618 PMCID: PMC11190877 DOI: 10.1021/acs.analchem.3c05557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/05/2024]
Abstract
Cerebral accumulation of amyloid-β (Aβ) initiates molecular and cellular cascades that lead to Alzheimer's disease (AD). However, amyloid deposition does not invariably lead to dementia. Amyloid-positive but cognitively unaffected (AP-CU) individuals present widespread amyloid pathology, suggesting that molecular signatures more complex than the total amyloid burden are required to better differentiate AD from AP-CU cases. Motivated by the essential role of Aβ and the key lipid involvement in AD pathogenesis, we applied multimodal mass spectrometry imaging (MSI) and machine learning (ML) to investigate amyloid plaque heterogeneity, regarding Aβ and lipid composition, in AP-CU versus AD brain samples at the single-plaque level. Instead of focusing on a population mean, our analytical approach allowed the investigation of large populations of plaques at the single-plaque level. We found that different (sub)populations of amyloid plaques, differing in Aβ and lipid composition, coexist in the brain samples studied. The integration of MSI data with ML-based feature extraction further revealed that plaque-associated gangliosides GM2 and GM1, as well as Aβ1-38, but not Aβ1-42, are relevant differentiators between the investigated pathologies. The pinpointed differences may guide further fundamental research investigating the role of amyloid plaque heterogeneity in AD pathogenesis/progression and may provide molecular clues for further development of emerging immunotherapies to effectively target toxic amyloid assemblies in AD therapy. Our study exemplifies how an integrative analytical strategy facilitates the unraveling of complex biochemical phenomena, advancing our understanding of AD from an analytical perspective and offering potential avenues for the refinement of diagnostic tools.
Collapse
Affiliation(s)
- Thomas Enzlein
- Center
for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack Str. 10, Mannheim 68163, Germany
- KU
Leuven-VIB Center for Brain & Disease Research, VIB, Leuven 3000, Belgium
- Department
of Neurosciences, Leuven Institute for Neuroscience and Disease, KU Leuven, Leuven 3000, Belgium
| | - Tammaryn Lashley
- Department
of Neurodegenerative Diseases, UCL Queen
Square Institute of Neurology, London WC1N 3BG, U.K.
| | - Denis Abu Sammour
- Center
for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack Str. 10, Mannheim 68163, Germany
| | - Carsten Hopf
- Center
for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack Str. 10, Mannheim 68163, Germany
- Mannheim
Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Heidelberg 69120, Germany
- Medical Faculty, Heidelberg University, Heidelberg 69120, Germany
| | - Lucía Chávez-Gutiérrez
- KU
Leuven-VIB Center for Brain & Disease Research, VIB, Leuven 3000, Belgium
- Department
of Neurosciences, Leuven Institute for Neuroscience and Disease, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
48
|
Shir D, Graff-Radford J, Fought AJ, Lesnick TG, Przybelski SA, Vassilaki M, Lowe VJ, Knopman DS, Machulda MM, Petersen RC, Jack CR, Mielke MM, Vemuri P. Complex relationships of socioeconomic status with vascular and Alzheimer's pathways on cognition. Neuroimage Clin 2024; 43:103634. [PMID: 38909419 PMCID: PMC11253683 DOI: 10.1016/j.nicl.2024.103634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION AD and CVD, which frequently co-occur, are leading causes of age-related cognitive decline. We assessed how demographic factors, socioeconomic status (SES) as indicated by education and occupation, vascular risk factors, and a range of biomarkers associated with both CVD (including white matter hyperintensities [WMH], diffusion MRI abnormalities, infarctions, and microbleeds) and AD (comprising amyloid-PET and tau-PET) collectively influence cognitive function. METHODS In this cross-sectional population study, structural equation models were utilized to understand these associations in 449 participants (mean age (SD) = 74.5 (8.4) years; 56% male; 7.5% cognitively impaired). RESULTS (1) Higher SES had a protective effect on cognition with mediation through the vascular pathway. (2) The effect of amyloid directly on cognition and through tau was 11-fold larger than the indirect effect of amyloid on cognition through WMH. (3) There is a significant effect of vascular risk on tau deposition. DISCUSSION The utilized biomarkers captured the impact of CVD and AD on cognition. The overall effect of vascular risk and SES on these biomarkers are complex and need further investigation.
Collapse
Affiliation(s)
- Dror Shir
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Angela J Fought
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Maria Vassilaki
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, 55905 USA
| | - Ronald C Petersen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Michelle M Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA; Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | | |
Collapse
|
49
|
de Lourdes Signorini-Souza I, Tureck LV, Batistela MS, Coutinho de Almeida R, Monteiro de Almeida S, Furtado-Alle L, Lehtonen Rodrigues Souza R. The potential of five c-miRNAs as serum biomarkers for Late-Onset Alzheimer's disease diagnosis: miR-10a-5p, miR-29b-2-5p, miR-125a-5p, miR-342-3p, and miR-708-5p. Brain Res 2024; 1841:149090. [PMID: 38880411 DOI: 10.1016/j.brainres.2024.149090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/29/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
The nervous system is rich in miRNAs, indicating an important role of these molecules in regulating processes associated with cognition, memory, and others. Therefore, qualitative and quantitative imbalances involving such miRNAs may be involved in dementia contexts, including Late-Onset Alzheimer's Disease (LOAD). To test the viability of circulating miRNAs (c-miRNAs) as biomarkers for LOAD, we proceed accordingly to the following reasoning. The first stage was to discover and identify profile of c-miRNAs by RNA sequencing (RNA-Seq). For this purpose, blood serum samples were used from LOAD patients (n = 5) and cognitively healthy elderly control group (CTRL_CH) (n = 5), all over 70 years old. We identified seven c-miRNAs differentially expressed (p ≤ 0.05) in the serum of LOAD patients compared to CTRL_CH (miR-10a-5p; miR-29b-2-5p; miR-125a-5p; miR-342-3p, miR-708-5p, miR-380-5p and miR-340-3p). Of these, five (p ≤ 0.01) were selected for in silico analysis (miR-10a-5p; miR-29b-2-5p; miR-125a-5p; miR-342-3p, miR-708-5p), for which 44 relevant target genes were found regulated by these c-miRNAs and related to LOAD. Through the analysis of these target genes in databases, it was possible to observe that they have functions related to the development and progress of LOAD, directly or indirectly connecting the different Alzheimer's pathways. Thus, this work found five promising serum c-miRNAs as options for biomarkers contributing to LOAD diagnosis. Our study shows the complex network between these molecules and LOAD, supporting the relevance of studies using c-miRNAs in dementia contexts.
Collapse
Affiliation(s)
- Isadora de Lourdes Signorini-Souza
- Postgraduate Program in Genetics. Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, 81531-990 Curitiba, Paraná State, Brazil
| | - Luciane Viater Tureck
- Postgraduate Program in Genetics. Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, 81531-990 Curitiba, Paraná State, Brazil
| | - Meire Silva Batistela
- Postgraduate Program in Genetics. Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, 81531-990 Curitiba, Paraná State, Brazil
| | - Rodrigo Coutinho de Almeida
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, LUMC, Leiden, the Netherlands
| | | | - Lupe Furtado-Alle
- Postgraduate Program in Genetics. Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, 81531-990 Curitiba, Paraná State, Brazil
| | - Ricardo Lehtonen Rodrigues Souza
- Postgraduate Program in Genetics. Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, 81531-990 Curitiba, Paraná State, Brazil.
| |
Collapse
|
50
|
Zhang Q, Yang G, Luo Y, Jiang L, Chi H, Tian G. Neuroinflammation in Alzheimer's disease: insights from peripheral immune cells. Immun Ageing 2024; 21:38. [PMID: 38877498 PMCID: PMC11177389 DOI: 10.1186/s12979-024-00445-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024]
Abstract
Alzheimer's disease (AD) is a serious brain disorder characterized by the presence of beta-amyloid plaques, tau pathology, inflammation, neurodegeneration, and cerebrovascular dysfunction. The presence of chronic neuroinflammation, breaches in the blood-brain barrier (BBB), and increased levels of inflammatory mediators are central to the pathogenesis of AD. These factors promote the penetration of immune cells into the brain, potentially exacerbating clinical symptoms and neuronal death in AD patients. While microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in AD, recent evidence suggests the infiltration of cerebral vessels and parenchyma by peripheral immune cells, including neutrophils, T lymphocytes, B lymphocytes, NK cells, and monocytes in AD. These cells participate in the regulation of immunity and inflammation, which is expected to play a huge role in future immunotherapy. Given the crucial role of peripheral immune cells in AD, this article seeks to offer a comprehensive overview of their contributions to neuroinflammation in the disease. Understanding the role of these cells in the neuroinflammatory response is vital for developing new diagnostic markers and therapeutic targets to enhance the diagnosis and treatment of AD patients.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Laboratory Medicine, Southwest Medical University, Luzhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, USA
| | - Yuan Luo
- Department of Laboratory Medicine, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China.
| | - Gang Tian
- Department of Laboratory Medicine, Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University, Sichuan, 646000, China.
| |
Collapse
|